Anda di halaman 1dari 63

TUGAS MATA KULIAH IMUNOLOGI

MEMBUAT JURNAL SALAH SATU PENYAKIT AUTOIMUN


“MYASTHENIA GRAVIS”

Nama Kelompok:
Annisa Widya Maharani (182210101010)
Erlik Fiana Sari (182210101013)
Lintang Qonita Fardliana (182210101016)
Wahyuni Ella Dwi Prasanti (182210101073)
Rika Ansyari (182210101138)

Nama Dosen: Ika Puspita Dewi, M.Biomed., Apt

FAKULTAS FARMASI
UNIVERSITAS JEMBER
2019
Annisa Widya Maharani 182210101010, Erlik gangguan ini disebut sindrom miastenia
Fiana Sari 182210101013 , Lintang Qonita kongenital. Myasthenia gravis adalah bentuk
Fardliana 182210101016, Wahyuni Ella Dwi kelompok penyakit terbesar dari gangguan
Prasanti 182210101073, Rika Ansyari 182210101138 neuromuscular junction dan disebabkan oleh
autoantibodi patogen ke komponen otot
postsynaptic. Fluktuasi keparahan
kelemahan otot yang khas. Beberapa, tetapi
Myasthenia Gravis tidak semua otot dipengaruhi dan tidak
Apa itu sistem kekebalan tubuh? selalu simetris. Peningkatan kelemahan
dengan aktivitas otot terus menerus
Sistem kekebalan tubuh adalah merupakan petunjuk diagnostik untuk
penyerangan atau pertahanan tubuh myasthenia gravis, namun gejala klinis dapat
terhadap infeksi atau bisa dikatakan sistem bervariasi. Pasien dengan myasthenia gravis
pada tubuh yang biasanya memproduksi harus diklasifikasikan menjadi
protein yang disebut “antibodi”. Antibodi subkelompok, supaya bisa ditentukan yang
berfungsi untuk membantu mencegah atau tepat untuk diagnosis, terapi yang optimal,
melawan infeksi. dan prognosis. Sebagai pengelompokan ini
memperhitungkan autoantibodi,
epidemiologi, klinis, dan komorbiditas.
Abstrak Autoantibodi terhadap reseptor asetilkolin
Dengan angka kejadian tahunan dari 8 (AChR), otot-spesifik kinase (Musk), dan
sampai 10 kasus per 1 juta orang, 4 lipoprotein protein 4 (LRP4) sebagai
myasthenia gravis dan berbagai penanda diagnostik yang sensitif dan
subkelompoknya adalah penyakit utama spesifik serta menjadi faktor pathogen.
yang mempengaruhi sambungan Autoantibodi ini juga sebagai salah satu
neuromuskuler. The Lambert- Eaton faktor untuk pengelompokan pasien
myasthenic sindrom dan neuromyotonia myasthenia gravis. (Gilhus dan
adalah presinaptik gangguan autoantibodi Verschuuren, 2015).
yang langka ditandai dengan disfungsi otot Myasthenia gravis adalah kondisi autoimun
rangka. 5 sindrom miastenia kongenital dan neuromuscular kronis yang menyebabkan
kondisi toksin (misalnya, botulism) dapat kelemahan otot rangka tubuh. MG
juga mempengaruhi sambungan disebabkan oleh kesalahan dalam transmisi
neuromuskuler dan menyebabkan impuls saraf ke otot.
kelemahan otot. Ulasan ini berfokus pada tes
baru diagnostik untuk myasthenia gravis dan Kelemahan tersebut dapat umum atau lokal,
individualisasi terapi sesuai dengan dan hampir selalu berdampak pada otot
biomarker (Havlícek, 1981). mata, dengan diplopia dan ptosis. Diplopia
adalah suatu gangguan penglihatan di mana
Disfungsi pada neuromuscular junction pasien akan melihat dua gambar dari satu
mendasari beberapa gangguan yang ditandai objek yang berdekatan (penglihatan ganda).
dengan kelemahan otot rangka yang Ptosis adalah istilah medis yang digunakan
melibatkan beberapa otot tapi tidak semua untuk menyatakan turunnya
kelompok otot. Bentuk genetik dari kelopak mata bagian atas sehingga tidak
dapat membuka mata sepenuhnya atau Apa yang menyebabkan Myasthenia
dalam bahasa sehari-hari kita sebut sebagai Gravis?
mata sipit sebelah. Pola keterlibatan
MG disebabkan oleh kesalahan dalam
gangguan mata biasanya simetris. Artinya
transmisi impuls saraf ke otot-otot. Pada
jika mata kanan mengalami gangguan
pasien MG, sistem kekebalan tubuhnya
seperti sipit sebelah, maka mata kiri akan
membuat antibodi yang keliru sehingga
mengikutinya. Kelemahan biasanya
menyerang koneksi antara saraf dan otot
meningkat dengan olahraga dan penggunaan
kemudian mencegah otot berkontraksi dan
otot berulang (kelelahan) dan bervariasi dari
mengakibatkan kelemahan. Beberapa orang
hari ke hari. (Havlícek, 1981)
mungkin memiliki Myasthenia Gravis yang
Myasthenia gravis merupakan penyakit tidak disebabkan oleh antibodi yang
autoimun yang berhubungan dengan menghalangi asetilkolin atau reseptor tirosin
antibodi terhadap reseptor asetilkolin, otot- kinase. Jenis kondisi ini disebut Myasthenia
spesifik reseptor tirosin kinase (Musk), Gravis Antibodi-Negatif. (Council for
terkait lipoprotein-protein 4 (LRP4), atau Medical Schemes, 2014)
Agrin dalam membran postsynaptic pada
Siapa yang mendapat Myasthenia
sambungan neuromuskuler. Pasien
Gravis?
myasthenia gravis harus diklasifikasikan
menjadi subkelompok untuk membantu Myasthenia Gravis terjadi pada semua
keputusan terapi dan prognosis. Pembagian kelompok etnis dan semua jenis kelamin. Ini
subkelompok didasarkan pada antibodi paling sering mempengaruhi wanita dewasa
serum dan awal gejala klinis, akhir-onset, muda (di bawah 40) dan pria yang lebih tua
thymoma, Musk, LRP4, antibodi-negatif, (lebih dari 60), tetapi juga dapat terjadi pada
dan bentuk mata dari penderita myasthenia usia berapa pun. Kondisi ini tidak diwarisi
gravis. Agrin terkait myasthenia gravis secara langsung juga tidak menular.
mungkin muncul sebagai entitas baru. (Council for Medical Schemes, 2014)
Prognosis baik dengan gejala optimal,
imunosupresif, dan pengobatan suportif.
Pyridostigmine adalah pengobatan Kasus Umum
simtomatik yang lebih disukai. Sementara
untuk pasien yang tidak merespon secara Miastema gravis adalah penyakit yang
memadai terapi simptomatik, kortikosteroid, jarang ditemui karena penyakit ini langka.
azatioprin, dan timektomi dianjurkan unruk Miestenia gravis biasanya terjadi pada usia
perawatan imunosupresif. Baru-baru ini obat 50 tahun keatas. Pada perempuan, penyakit
imunomodulator tambahan ditemukan, tetapi ini biasanya muncul di usia sektar 30 tahun
keputusan terapeutik terhambat oleh sedangkan pada laki laki, penyakit ini
kelangkaan studi terkontrol. Terapi obat muncul di usia sekitar 60 tahun. Kondisi ini
jangka panjang penting untuk sebagian besar disebabkan oleh gangguan komunikasi
pasien dan harus disesuaikan dengan bentuk antara saraf dan otot yang mana biasanya
myasthenia gravis tertentu. (Gilhus dan terjadi kelemahan pada otot lengan dan kaki,
Verschuuren, 2015) pandangan bias, serta kesulitan untuk
berbicara. Kondisi ini dapat bertaha selama
bertahun tahun hingga seumur hidup.
Anatomi dan Fisiologi Neomuscular terjadinya depolarisasi di post sinaps.
junction Kontraksi yang terjadi yaitu akibat dari
potensial aksi pada sel otot tersebut. Enzym
Neomuscular junction atau synaptic
juga berperan untuk menghidrolisis Ach
transmission adalah gangguan yang
yang masih tertempel pada AchR. Sehingga
menyebabkan salah satu karakteristik akibat
untuk mencegah terjadinya kontraksi yang
dari Miastemia gravis. Terdapat potensial
terus menerus yaitu dengan Asetilkolin yang
aksi pada neuron motorik dimana sewaktu
akan dipecah menjadi 2 yaitu Kolin dan
mendekati otot, akson membentuk cabang.
Asam Laktat, dimana Kolin akan masuk
Dari cabang cabang tersebut akan
kembali kedalam membran pre sinaps dan
membentuk otot secara keseluruhan yang
membentuk Asetillkolin kembali.
mana bentuknya panjang dan silinder. Pada
Neomuscular junction ini sel otot dan sel Patofisiologi
saraf tidak berikatan melainkan terdapat
Asetilkolin yang dilepaskan dalam jumlah
celah antara keduanya. Celah yang ada pada
yang normal, tidak akan memberikan efek
sel otot dan sel saraf ini sangat besar
potensial aksi dimana efek potensial aksi
sehingga memungkinkan terjadinya
tersebut tidak akan sampai ke membran post
transmisi suatu impuls diantara sel saraf dan
sinaps yang mana pada kondisi Miastenia
sel otot pada Neomuscular junction tersebut.
gravis ini terjadi penurunan jumlah Ach-R.
Sama seperti hal nya pada sinaps, yaitu
Sehingga ketika kekurangan reseptor
terdapat pembawa informasi dimana
tersebut, akan mengakibatkan penurunan
menyambungkan antara sel saraf dan sel
jumlah yang akan diaktifkan oleh saraf dan
otot, maka dari itu neurotransmitter disini
otot tertentu. Hal hal ini lah yang
juga disebut sebagai Ach (Asetilkolin).
menyebabkan pasien merasa sakit, yaitu
Vesikel berfungsi untuk menyimpan karena kekurangan reseptor dan keberadaan
membran pre sinaptik yang mengandung Ach dalam jumlah normal. Antibodi pada
Asetilkolin dimana apabila terbukanya reseptor asetilkolin itu adalah penyebab
channel Ca+ Voltage Gated akan utama kelemahan otot pada penyakit
menyebabkan influks kalsium. Influks Miestenia gravis. Pada beberapa orang
kalsium ini yaitu masuknya kalsium pada mungkin memiliki Miestenia gravis yang
membran dimana vesikel yang diaktifkan tidak disebabkan oleh antibodi yang
oleh influks tadi akan mendocking ke tepi menghalangi asetilkolin, maka dari itu
membran. Sehingga setilkolin atau Ach tadi kondisi yang seperti ini yaitu dimana tidak
akan dilepaskan ke celah sinaptik. Ach yang disebabkan oleh antibodi yang menghalangi
telah dilepaskan ke celah sinaptik tadi akan asetilkolin yaitu disebut sebagai antibodi-
berikatan dengan AchR yang terdapat di negatif Miestenia gravis. Pada kelenjar
membran post sinaps. timus juga memiliki peran untuk mendeteksi
adanya Miestenia gravis, yaitu dengan
Asetilkolin diikat oleh 5 sub-unit protein
membesarnya timus dimana hal tersebut
dari Ach-R dimana akan membentuk sebuah
tidak normal.
lingkaran. Ikatan AchR dengan Ach
(Asetilkolin) akan menyebabkan influks
Na+ dimana Na+ ini akan mengakibatkan
Epidemologi Kondisi tersebut dapat disertai kelumpuhan
anggota badan terutama triceps dan
Pada penyakit Miastenia gravis ini terjadi
ekstensor jari-jari. kelemahan/kelumpuhan
pada usia muda yang mana sekitar 30tahun
otot lainnya dapat terjadi pada otot
untuk wanita dan 60 tahun untuk pria.
pernafasan sehingga dapat menyebabkan
Miastenia gravis ini juga terjadi dari 20
sesak nafas.
dalam 100.000 populasi yang mana sangat
jarang terjadi. Insiden puncak di umur yang 2. Tes klinik sederhana
lebih awal pada wanita dibandingkan dengan
a). Tes watenberg/simpson test : Pasien
laki laki yaitu karena wanita memiliki
akan diminta untuk melihat objek yang
gangguan imun yang lebih banyak
diletakkan diantara kedua mata pasien.
dibandingkan pria. Sehingga, Miastenia
Apabila dalam kurun waktu > 30 detik mata
gravis memiliki onset yang lebih sering pada
pasien mengalami ptosis (kelopak mata
wanita dibandingkan dengan laki laki. Di
turun sebelah) maka tes dinyatakan positif.
Asia Timur, 50% dari semua kasus memiliki
onset sebelum usia 15tahun pada b). Tes pita suara : Pada tes ini, biasanya
subkelompok mata. Dimana antibodi LRP4 pasien akan diminta untuk menghitung 1-
atau lipoprotein yang terkait dengan protein 100. Apabila suara pasien semakin lama
ini terdapat di pasien tanpa antibodi Ach-R. semakin menghilang maka tes akan
Data epidemologi bahwasanya LRP4 yang dinyatakan positif .
berkaitan dengan Miastenia gravis ini terkait
dengan musk. Tetapi hal ini lebih sering 3. Uji Tensilon (edrophonium chloride)
terjadi pada orang orang yang ada di Eropa. uji tensilon dilakukan dengan menyuntikkan
Faktor yang menyebabkan yaitu bisa dari 2 mg tensilon secara intravena selama 15
luar seperti diet atau infeksi. detik, apabila dalam 30 detik tidak terdapat
reaksi maka volume penyuntikan dinaikkan
sebanyak 8-9 mg tensilon. Setelah tensilon
DIAGNOSIS MIASTENIA GRAVIS disuntikkan, perhatikan otot-otot yang
mengalami pelemahan, biasanya terjadi
Diagnosis untuk penyakit miastenia
pada kelopak mata yang memperlihatkan
gravis diperlukan pengujian yang bersifat
adanya ptosis. Apabila kelemahan otot mata
akurat. Diagnosis penyakit ini dapat
disebabkan oleh Miastenia gravis, maka
ditegakkan berdasarkan beberapa pengujian
ptosis akan pulih. Pada uji ini kelemahan
seperti anamnesis, tes antikolinesterase,
otot mata harus diperhatikan dengan sangat
EMG, serologi untuk antibodi AchR dan
seksama, karena efektivitas dari tensilon
CT-Scan atau MRI toraks untuk melihat
sangat singkat. Efek sampingnya dapat
adanya timoma.
menyebabkan bradikardi dan untuk
1. Anamnesis mengatasinya dapat digunakan atropin.
Anamnesis merupakan kelemahan atau
kelumpuhan otot yang berulang setelah
aktivitas dan membaik setelah istirahat.
Biasanya miastenia gravis menyerang otot
mata dengan manifestasi: diplopi atau ptosis.
neuromuskular fiber berupa adanya
peningkatan titer dan fiber pada densitas
4. Uji Prostigmin (neostigmin)
normal. Pengujian teknik ini menggunakan
Pada uji prostigmin dilakukan jarum single-fiber yang memiliki permukaan
penyuntikkan 3 cc atau 1,5 mg prostigmin kecil untuk merekam serat otot penderita,
metilsulfat secara intramuscular sehingga SFEMG dapat mendeteksi suatu
(ditambahkan atropin 0,8 mg apabila titer (variabilitas pada interval interpotensial
diperlukan). Jika kelemahan otot diantara 2 atau lebih serat otot tunggal pada
disebabkan oleh Miastenia gravis maka unit yang sama) dan fiber density (jumlah
gejala seperti ptosis, strabismus atau potensial aksi dari serat otot tunggal yang
kelemahan lain tidak lama kemudian akan dapat direkam oleh jarum perekam).
membaik. Pada pasien tanpa timoma anti- Repetitive Nerve Stimulation (RNS)
muscle-specific antibodi dapat menunjukkan mendeteksi adanya penurunan jumlah
hasil positif pada pasien dengan usia lebih reseptor asetilkolin pada penderita Miastenia
dari 40 tahun. gravis, sehingga pada RNS terdapat
penurunan potensial aksi.
Pada Anti-musclespecific kinase
(MuSK), hampir 50% antibodi penderita 7. Gambaran Radiologi
Miastenia gravis yang menunjukkan hasil
Chest x-ray (foto roentgen thorak)
anti-AChR Ab negatif (Miastenia gravis
dapat dilakukan dalam posisi anteroposterior
seronegatif) dan hasil positif ditunjukkan
dan lateral. Pada roentgen thorak, thymoma
dari anti-MuSK Ab. Kemudian Anti-
dapat diidentifikasi sebagai suatu massa
asetilkolin reseptor antibodi, hasil dari
pada bagian anterior mediastinum. Pada
pemeriksaan dapat digunakan untuk
hasil roentgen negative, belum dipastikan
mendiagnosis suatu Miastenia gravis,
dapat menyingkirkan adanya thymoma
dimana hasil yang postif ditunjukkan pada
berukuran kecil. Sehingga perlu dilakukan
70% - 95% dari penderita Miastenia gravis
chest Ct-scan untuk mengidentifikasi
generalisata dan 50% - 75 % dari penderita
thymoma pada seluruh kasus Miastenia
dengan Miastenia okular murni
gravis, terutama pada penderita dengan usia
menunjukkan hasil tes anti-asetilkolin
tua. Pemerikasaan MRI pada otak dan orbita
reseptor antibodi positif. Pada pasien
sebaiknya tidak digunakan sebagai
timoma tanpa mengidap Miastenia gravis
pemeriksaan rutin. Namun, MRI dapat
sering kali terjadi false positive anti-AChR
digunakan apabila diagnosis Miastenia
antibody.
gravis tidak dapat ditegakkan dengan
6. Elektrodiagnostik pemeriksaan lainnya serta mencari
penyebab defisit pada saraf otak.
Pemeriksaan elektrodiagnostik dapat
memperlihatkan adanya kerusakan pada Autoimun adalah kondisi ketika system
transmisi neuromuskular melalui 2 teknik kekebalan tubuh mengalami kelainan
yaitu Single-fiber Electromyography sehingga menyerang jaringan dan saraf yang
(SFEMG) dan Repetitive Nerve Stimulation sehat pada tubuh. Penyakit myasthenia
(RNS). Pada pengujian SFEMG mendeteksi gravis ini memiliki banyak gejala. Berikut
adanya kerusakan transmisi pada gejala pada myasthenia gravis.
Ada beberapa gejala yang terjadi pada tersebut tidak ada atau hilang ketika salah
penyakit myasthenia gravis diantarannya: satu mata ditutup. Biasanya pada diplopia
1. Gejala yg utama yaitu melemahnya bonikular ini ditandai dengan pergerakan
otot otot kerangka misalnya otot otot bola mata terganggu sehingga sudut
wajah, mata, tenggorokan dan kaki. mata nya tidak sinkron.

Gejala ini terjadi setelah beraktivitas  Diplopia monocular :


dan hilang setelah istirahat. Biasanya
Gejala ini jika salah satu mata
otot melemah itu terjadi ketika otot kita
ditutup, maka akan terjadi buram sebelah.
tidak berkontraksi secara normal.
Biasanya terjadi pada penderita katarak,
Contoh saja pada otot kaki. Biasanya
kornea , lensa mata dan gangguan pada
kita sulit untuk bergerak ketika kita
retina.
duduk kemudian berdiri, pincang dan
kesulitan mengangkat barang. 4. Kesulitan mengunyah dan menelan
makanan
2. Ptosis (kelopak mata droopy)
Gejala ini ditandai adanya nyeri pada
Kondisi ini biasanya disebabkan
peradangan mulut sehingga menyebabkan
karena trauma, usia, dan gangguan
kesulitan untuk mengunyah dan menelan
kesehatan. Biasannya gejala ini ditandai
makanan.
turunya kelopak mata bagian atas
sehingga matanya tidak bisa membuka
sepenuhnya atau sempurna, dan 5. Kesulitan berbicara
biasanya sipit sebelah. Selain itu pada
ptosis dapat terjadi pada kedua mata Gejala ini ditandai ketika berbicara
atau bilateral. Kondisi ini menjadikan tidak seperti biasanya. Suara rendah atau
mata berair sehingga dapat menganggu kecil seperti hidungnya tersumbat atau
penglihatan kita. pengap. sehingga cara berbicaranya itu
tidak bisa lancar.
3. Diplopia (penglihatan kabur atau
ganda) 6. Kesulitan bernafas
Gejala ini biasanya kita melihat
Gejala ini ditandai pada Kelemahan
sesuatu itu tidak persis dengan arah yang
otot pernafasan yang menyebabkan
sama atau bersamaan. Jadi kita bisa melihat
penderita sulit untuk bernafas. Dada terasa
sesuatu yang ada bayangannya. Gejala ini
berat untuk bernafas bahkan bisa terjadi
ditandai dengan kelelahan dan banyaknya
sesak nafas atau karena melemahnya otot
mengkonsumsi alkohol.
otot dinding dada.
Diplopia secara umum dibagi menjadi 2 7. Kehilangan ekspresi wajah
yaitu:
Gejala ini ditandai dengan otot wajah
 Diplopia bonikular : melemah sehingga kelihatan sulit untuk
menunjukkan ekspresi wajah misalnya
biasanya penderita melihat objek tersenyum.
dengan kedua mata dan kemudian objek
Myasthenia gravis juga ditandai terjadi pada penyakit myasthenia gravis
dengan cepat kelelahan setelah tidak bisa dibuat kerja yang berat atau
menggerakkan otot. Biasanya penderita beraktivitas terlalu lama.
penyakit myasthenia ini memiliki tanda dan
gejala yang berbeda beda. Jika gejala yang Algoritme terapi pada myasthenia gravis
terjadi pada tubuh kita tidak langsung Myasthenia gravis merupakan penyakit yang
ditangani, maka gejala tersebut bisa berkepanjangan (kronis) dan cenderung
berkembang semakin parah. Biasanya, memburuk seiring berjalannya waktu.
gejala pada myasthenia gravis semakin Penderita myasthenia gravis perlu
memburuk jika kita melakukan banyaknya melakukan pemeriksaan dokter secara
aktivitas. berkala agar perkembangan penyakit dan
kondisinya dapat terpantau dengan baik.
Kondisi melemahnya otot akibat Penderita myasthenia gravis dianjurkan
penyakit myasthenia gravis juga dapat untuk segera ke IGD bila mengalami sesak
menyerang bagian tubuh yang lain, seperti napas. Kondisi ini dapat berkembang
otot leher, lengan dan tungkai. Biasanya menjadi henti napas, sehingga penderita
gejala yang sering muncul yaitu : perlu mendapatkan alat bantu pernapasan
secepatnya.Walaupun belum ada cara yang
 Nyeri otot setelah beraktivitas efektif untuk menyembuhkan myasthenia
 Sulit mengangkat kepala setelah gravis, tetapi pengobatan yang diberikan
berbaring oleh dokter dapat meredakan gejala,
 Sulit bergerak, naik turun tangga. meningkatkan fungsi otot, dan mencegah
kelumpuhan otot-otot pernapasan yang
 Dan gangguan dalam berjalan
berakibat fatal.Jenis penanganannya pun
berbeda-beda untuk tiap penderita,
Dalam kasus myasthenia gravis, tergantung usia, tingkat keparahan, dan
antibody menghalangi , menyerang dan kondisi pasien secara keseluruhan.
menghancurkan reseptor asetilkolin yang
dibutuhkasn otot untuk bisa berkontraksi. Beberapa tindakan pengobatan untuk
mengatasi myasthenia gravis adalah:
Jadi melemahnya otot yang ada pada tubuh
itu menyebabkan otot tidak bisa A. Acetilkolinesterase inhibitor dapat
berkontraksi. Penyakit myanesthesia gravis diberikan piridostigmin bromida
ini sangat berbahaya karena bisa (mestinon) 30-120 mg/3-4 jam/oral.
menyebabkan kerusakan pada otot saraf dan Dosis parenteral 3-6 mg/4-6 jam/ iv tiap
otot yang lainnya. hari akan membantu pasien untuk
Dari gejala yang dialami oleh penderita mengunyah, menelan, dan beberapa
aktivitas sehari-hari. Pada malam hari,
penyakit myasthenia gravis biasanya
dapat diberikan mestinon long-acting 180
sembuhnya lama, karena dari gejala tersebut
mg. Apabila diperlukan, neostigmin
bisa menyebabkan otot otot dalam tubuh bromida (prostigmine ): 7,5-45 mg/2-6
melemah dan bisa sembuh jika penderita jam/oral. Dosis parenteral : 0,5-1 mg/4
menghindari atau tidak melakukan banyak jam/iv atau im. Neostigmin dapat
aktivitas dan perlu diwaspadai jika gejala menginaktifkan atau menghancurkan
tersebut semakin parah maka segera kolinesterase sehingga asetilkolin tidak
ditangani secepatnya karena gejala yang segera dihancurkan. Akibatnya aktifitas
otot dapat dipulihkan mendekati normal, PE paling efektif digunakan pada situasi
sedikitnya 80-90% dari kekuatan dan dimana terapi jangka pendek yang
daya tahan semula. Pemberian menguntungkan menjadi prioritas.Dasar
antikolinesterase akan sangat bermanfaat terapi dengan PE adalah pemindahan anti-
pada Miastenia gravis golongan IIA dan asetilkolin secara efektif.Respon dari terapi
IIB. Efek samping pemberian ini adalah menurunnya titer antibodi.
antikolinesterase disebabkan oleh Dimana pasien yang mendapat tindakan
stimulasi parasimpatis, termasuk berupa hospitalisasi dan intubasi dalam
konstriksi pupil, kolik, diare, salivasi waktu yang lama serta trakeostomi, dapat
berkebihan, berkeringat, lakrimasi, dan diminimalisasikan karena efek dramatis dari
sekresi bronkial berlebihan. Efek PE.Terapi ini digunakan pada pasien yang
samping gastro intestinal (efek samping akan memasuki atau sedang mengalami
muskarinik) berupa kram atau diare dapat masa krisis. PE dapat memaksimalkan
diatasi dengan pemberian propantelin tenaga pasien yang akan menjalani
bromida atau atropin. 2, 8, 10, 12 timektomi atau pasien yang kesulitan
menjalani periode pasca operasi. Jumlah dan
B. Kortikosteroid volume dari penggantian yang dibutuhkan
Dapat diberikan prednison dimulai dengan kadang-kadang berbeda tetapi umumnya 3-4
dosis rawal 10-20 mg, dinaikkan bertahap liter sebanyak 5x dalam 2 minggu. 4, 10, 12
(5-10 mg/minggu) 1x sehari selang sehari, E. Intravenous Immunoglobulin (IVIG)
maksimal 120 mg/6 jam/oral, kemudian
diturunkan sampai dosis minimal efektif. Dosis standar IVIG adalah 400 mg/kgbb/hari
Efek sampingnya dapat berupa: peningkatan pada 5 hari pertama, dilanjutkan 1
gram/kgbb/hari selama 2 hari. 2, 10, 12
berat badan, hiperglikemia, osteopenia,
ulkus gaster dan duodenum, katarak. 2, 8, F. Timektomi
10, 12
Timektomi umumnya dianjurkan pada
C. Azatioprin pasien umur 10-55 tahun dengan Miastenia
gravis generalisata. Walaupun timektomi
Azatioprin merupakan suatu obat merupakan terapi standar di berbagai pusat
imunosupresif, juga memberikan hasil yang pengobatan namun keeefektivitasannya
baik, efek sampingnya sedikit jika belum dapat dipastikan oleh penelitian
dibandingkan dengan steroid dan terutama prospektif yang terkontrol. Timektomi
berupa gangguan saluran cerna,peningkatan diindikasi pada terapi awal pasien dengan
enzim hati, dan leukopenia. Obat ini keterlibatan ekstremitas bawah dan bulbar
diberikan dengan dosis 2-3 mg/kg
BB/hari/oral selama 8 minggu pertama.
Setiap minggu harus dilakukan pemeriksaan
Jika berat firsline dipakai terapi golongan
darah
acetylcholinesterase yaitu piridostigmin atau
lengkap dan fungsi hati. Sesudah itu mestinon ,jika terapi berhasil maka
pemeriksaan laboratorium dikerjakan setiap dilanjutkan jika tidak diganti dengan
bulan sekali. Pemberian prednisolon prednisone atau azathioprine jika berhasil
bersama-sama dengan azatioprin sangat dilanjutkan dengan dosis rendah,jika tidak
dianjurkan. 2, 12
berhasil diganti sesuai dengan kondisi
D. Plasma Exchange (PE) sebagai berikut :
Jika kondisi ringan sampai sedang memakai
terapi berupa micofenolat jika berat
memakai rituximab jika berhasil dilanjutkan
tetapi jika tidak berhasil diganti dengan
imunosupresan lain seperti;
tacrolimus,methotrexate,azathioprine

Referensi
Conti-Fine BM, Milani,Monica
,Kaminski,Henry J. . Myasthenia gravis:
past, present, and future. The Journal of
Clinical Investigation 2006;116(Number 11)
Council for Medical Schemes. 2014. CM s
cript. CM S Cript. (4)
Gilhus, N. E. dan J. J. Verschuuren. 2015.
Myasthenia gravis: subgroup
classification and therapeutic strategies.
The Lancet Neurology. 14(10):1023–
1036.
Havlícek, F. 1981. Myasthenia gravis.
Casopis Lekaru Ceskych.
120(36):1073–1077.
Ropper AH, Brown, Robert H. ,. Adam And
Victor's Principles of Neurology. In:
Myasthenia Gravis And Related Disorders
Of The Neuromuscular Junction 8 th ed.
United State of America: McGraw-Hill
Medical Publishing Division; 2005.
Setiyohadi B. Miologi. In: Sudoyo AW,
Setiyohadi, Bambang, Alwi, idrus,
Simadibrata K.,Marcellus, Setiati, Siti,
editor. Buku Ajar Ilmu Penyakit Dalam.
Jakarta: InternaPublishing; 2009.
Lampiran Jurnal Referensi

Myasthenia gravis: subgroup classification and


therapeutic strategies
Nils Erik Gilhus, Jan J Verschuuren

Myasthenia gravis is an autoimmune disease that is characterised by muscle weakness and Lancet Neurol 2015; 14:
fatigue, is B-cell mediated, and is associated with antibodies directed against the 1023–36
acetylcholine receptor, muscle-specific kinase (MUSK), lipoprotein-related protein 4 (LRP4),
Department of Clinical
or agrin in the postsynaptic membrane at the neuromuscular junction. Patients with Medicine, University of
myasthenia gravis should be classified into subgroups to help with therapeutic decisions Bergen,
and prognosis. Subgroups based on serum antibodies and clinical features include early- Bergen, Norway
onset, late-onset, thymoma, MUSK, LRP4, antibody-negative, and ocular forms of myasthenia (Prof N E Gilhus MD);
Department of Neurology,
gravis. Agrin-associated myasthenia gravis might emerge as a new entity. The prognosis is Haukeland University
good with optimum symptomatic, immunosuppressive, and supportive treatment. Hospital,
Pyridostigmine is the preferred symptomatic treatment, and for patients who do not Bergen, Norway
adequately respond to symptomatic therapy, corticosteroids, azathioprine, and thymectomy (Prof N E Gilhus); and
Department of
are first-line immunosuppressive treatments. Additional immunomodulatory drugs are Neurology, Leiden
emerging, but therapeutic decisions are hampered by the scarcity of controlled studies. University Medical
Long-term drug treatment is essential for most patients and must be tailored to the particular Center, Leiden,
form of myasthenia gravis. Netherlands (Prof J J
Verschuuren MD)
Correspondence to:
Prof Nils Erik Gilhus,
Department

Introduction clinical features can vary. Patients with instrumental for


Dysfunction at the neuromuscular junction myasthenia gravis should be classified into subgrouping
underlies several disorders that are subgroups, with implications for diagnosis, patients with
characterised by skeletal muscle weakness optimum therapy, and prognosis. In myasthenia
usually involving some but not all muscle myasthenia gravis guidelines and consensus gravis. A
groups. Genetic forms of these disorders are reports, subgrouping is recommended,1–5 but
termed congenital myasthenic syndromes. exact definitions vary and new subgroups are
Some toxins, like botulinum toxin and curare, emerging as a result of increased knowledge.
can cause neuromuscular dysfunction; As this subgrouping takes into account
acquired antibody-mediated forms include myasthenia gravis autoantibodies,
autoimmune and neonatal myasthenia gravis, epidemiology, clinical presentation, and
Lambert– Eaton myasthenic syndrome, and comorbidities, the subgroups are discussed
neuromyotonia. after these sections in this Review. For a few
Myasthenia gravis forms the largest disease patients, subgrouping is not possible owing to
group of neuromuscular junction disorders and insufficient precise information, including
is caused by pathogenic autoantibodies to suboptimum autoantibody testing and
components of the postsynaptic muscle pathological changes of the thymus below the
endplate (figure 1).1–4 Fluctuations in severity of detection threshold of imaging.
muscle weakness are typical. Some, but not all, Autoantibodies against the acetylcholine
muscles are affected and not necessarily receptor (AChR), muscle-specific kinase
symmetrically. Increased weakness with (MUSK), and lipoprotein- related protein 4
continued muscle activity represents a (LRP4) are well established as sensitive and
diagnostic clue for myasthenia gravis, but these specific diagnostic markers and pathogenic
factors, and these autoantibodies are
prerequisite for optimum diagnosis and of Neurology, Haukeland University Hospital, 5021 Bergen,
Norway
treatment, therefore, is access to autoantibody
nils.gilhus@helse-bergen.no
testing.1–5
With modern immunosuppressive,
symptomatic, and supportive treatments, the
prognosis for patients with myasthenia gravis is
good. Most patients with mild-to- moderate
symptoms will obtain full remission or
substantial improvement. Full remission is rare
in severe cases, some variation over time is
common, and steady progression is unusual.
Daily life functions of individuals with
myasthenia gravis are not, or only modestly,
affected and life expectancy is not reduced.6
Long-term drug treatment is necessary for
nearly all patients with myasthenia gravis.2,7 In
10–15% of these patients, full control of the
disease is not possible or is only at the cost of
severe side-effects of immunosuppressive
therapy.8
Treatment protocols at leading centres are
not based purely on results from well
controlled studies or guidelines based on such
studies, because well controlled studies are
sparse for this disease, and do not take into
account the variation in therapeutic response
among the diagnostic subgroups. Myasthenia
gravis is a rare disease, and most patients do
well on existing treatments, both aspects that
are a challenge for new trials. We will combine
information from controlled studies, consensus
reports, and expert views with insights from
theoretical and experimental studies relevant
for myasthenia gravis subgroups, with the aim
of assessing the evidence base for the use of
treatments, including interventions directed at
the patho- physiological process.

Autoantibodies in myasthenia gravis


AChR antibodies are highly specific for
myasthenia gravis, and their presence
combined with muscle weakness confirms the
disease. Further diagnostic investigation is
necessary only to define the subgroup and
disease severity. The value of repeated AChR
antibody testing in patients with this disorder is
debated,
A Standard tests for MUSK antibodies use radio-
immunoprecipitation or an ELISA. Cell-based assays
Schwann cell
used for research can increase sensitivity. MUSK
Nerve antibodies are directly pathogenic in experimental
animal models,11–13 even if the predominant IgG4
antibodies do not bind complement. Any value of
ACh Vesicles repeated tests in the follow-up of patients has not been
established because prospective, high-quality studies
have not been done.
LRP4 antibodies bind to the membrane protein in
vivo, block the agrin–LRP4 interaction and thereby also
inhibit AChR clustering in the membrane. Interference
with the LRP4–MUSK interaction might also be a
Muscle relevant disease mechanism for this subgroup. Mice
immunised with LRP4 develop typical myasthenia
gravis.14 Thus, LRP4 antibodies are directly pathogenic
through interference with AChR function.
Agrin antibodies have been detected in a few patients
with myasthenia gravis and AChR, MUSK, or LRP4
antibodies.15,16 Agrin is essential for AChR function, but
Agrin Acetylcholin
B C e AChR
whether these antibodies contribute to the muscle
LRP4–MUSK
complex weakness in this disease is still unclear. Similarly,
Nerv Nerv cortactin autoantibodies have been reported in patients
e e with myasthenia gravis, both with and without other
Synaps Synaps
e
neuromuscular autoantibodies.17
e
Titin and ryanodine receptor antibodies occur in
some patients with AChR-associated myasthenia gravis.
Titin maintains the flexibility of the cell structure,
whereas the ryanodine receptor is a sarcoplasmic
reticulum calcium channel that mediates contraction of
Muscl Muscl the muscle cell. Titin and ryanodine receptor antibodies
e e
probably do not enter the muscle cell in vivo and might
Figure 1: The neuromuscular junction not mediate any muscle weakness, but rather could be
(A) The AChR and MUSK are expressed at the top of the junctional folds. (B) Trophic signal: binding of agrin disease markers.18 These antibodies are present with a
to the LRP4–MUSK complex activates aggregation of AChRs and promotes transition from the plaque to pretzel high frequency in thymoma- associated myasthenia
form of the neuromuscular junction. (C) Activation signal: binding of acetylcholine to the AChR induces a gravis, with an intermediate frequency in late-onset
brief opening of the central ion channel causing membrane depolarisation, which in turn elicits a muscle action
potential that leads to contraction of the muscle fibre. AChR=acetylcholine receptor. MUSK=muscle-specific
myasthenia gravis, and very rarely in early-onset and
kinase. ocular myasthenia gravis; they are not detected by
LRP4=lipoprotein-related protein 4. standard testing in MUSK, LRP4, or antibody- negative
myasthenia gravis.7,19 Titin and ryanodine receptor
but changes in antibody concentration might predict antibodies can be used to diagnose a thymoma in
disease severity in patients given immunosuppressive patients younger than 50 years. 19 These antibodies have
drugs and therefore can support therapeutic decisions. been proposed as markers for severe myasthenia gravis
No correlation has been shown between AChR antibody with a need for long-term immunosuppression and no
concentration and disease severity. AChR antibodies response to thymectomy. Commercial tests with ELISA
are directly pathogenic through crosslinking of AChRs are available for titin but not for ryanodine receptor
leading to accelerated degradation of these receptors, antibodies.
through complement binding and activation, and by
inducing AChR conformational changes or blocking Epidemiology
acetylcholine binding.1–4 Radioimmunoprecipitation is Autoimmune myasthenia gravis has a reported
the standard commercial test and gives a quantitative worldwide prevalence of 40–180 per million people,
AChR antibody measure. Cell-based assays can have an and an annual incidence of 4–12 per million people. 20–23
even higher sensitivity than radioimmunoprecipitation, Recently collected figures of prevalence and incidence
but are not yet commercially available and tend to be higher than older ones, especially for late-
standardised.9 Tests avoiding radioactive ligands are onset myasthenia gravis, partly explained by increased
also in use such as ELISA and fluorescence tests based case finding and more widespread autoantibody
on immuno- precipitation, 10 but they tend to be less testing. Population demographics with an increased
sensitive than assays with radioactive ligands. number of elderly people and reduced myasthenia
gravis mortality affect incidence and prevalence. AChR-
associated
myasthenia gravis has a bimodal age pattern of
incidence, with a peak in young adults aged about SubtypeAChRMUSKLRP4 myasthenia gravis myasthenia gravis Seronegative LEMS
30 years and then a steady increase in incidence with myasthenia gravis myasthenia
gravis
increasing age older than 50 years. 20,21 The incidence
peak in young adults is mainly because of the high
frequency in women, typical for many autoimmune
disorders, although late-onset myasthenia gravis is
slightly more frequent in men. No evidence suggests
that the occurrence of this disease is increasing as a
result of a change in external causative factors such as
infections or diet.24 Relative prevalence 80% 4% 2% 5% 4%
Overall, myasthenia gravis incidence and prevalence
shows little geographical variation; however, this
Figure 2: Distribution of weakness and relative prevalence of subtypes of myasthenia
distribution is not the case for all subgroups of the gravis AChR=acetylcholine receptor. MUSK=muscle-specific kinase. LRP4=lipoprotein-related
disease. Juvenile myasthenia gravis, a subtype of early- protein 4. LEMS=Lambert–Eaton myasthenic syndrome.
onset disease, has a high frequency in east Asia, in which
up to 50% of all cases have onset before age 15 years, Comorbidities
many of them with ocular symptoms only.22,25 Myasthenia Patients with early-onset and ocular subgroups of
gravis incidence in children (aged <15 years) in a mixed myasthenia gravis have increased frequency of organ-
population from Canada was 1–2 per million per year, specific and general autoimmune disorders, especially
and highest in those of Asian ethnicity, especially for the thyroiditis.29 Patients with thymoma-associated
ocular subgroup. LRP4 antibodies were recorded in 19% myasthenia gravis are at an increased risk of
of patients without AChR antibodies,5 and MUSK developing haematological autoimmune disorders.
antibodies in a third of patients without AChR Thymectomies have not been shown to increase the
antibodies.3,4,26 Epidemiological data suggest that LRP4- risk of infections, autoimmune disease, or cancer.
associated myasthenia gravis is half as frequent as the Myasthenia gravis muscle weakness might increase the
MUSK form of the disease. MUSK-associated myasthenia risk of respiratory infections and osteoporosis,
gravis incidence is estimated at 0·3 patients per million becoming overweight, and developing other
per year, with a prevalence of 2·9 per million people, and complications. A widespread autoimmune
is more common in southern than northern Europe.27 inflammatory myopathy can occur in myasthenia
Genetic predisposition and external factors linked to gravis.30 AChR antibodies and myasthenia gravis-like
infections or diet are potential explanations for some features have been described occasionally in patients
geographical variation in this disease and its subtypes. with amyotrophic lateral sclerosis.31
Several studies32–34 have investigated the cancer risk in
Clinical presentation patients with myasthenia gravis and its subgroups.
Muscle weakness is a major symptom and sign in Methodological challenges due to myasthenia gravis
myasthenia gravis. The combination of weakness patient selection, sensitivity in cancer detection, follow-up
localisation, variation in weakness over time, and time, and types of control groups have led to varying
exercise- induced weakness usually gives strong clues to conclusions. Thymomas in general seem to confer a
the diagnosis of the disease for all subgroups. In older moderately increased risk for other cancer types, 32
individuals with eye muscle weakness and bulbar whereas myasthenia gravis and its immunoactive
symptoms, cerebrovascular disease of the brainstem is treatment, according to a Danish population-based study 33
sometimes suspected. In younger individuals, unspecific with a long-term follow-up and relevant controls, was
fatigue disorders can be part of the differential not associated with a significantly increased risk,
diagnoses.1,3,7 Weakness in myasthenia gravis arises in the perhaps with the exception of non-melanoma skin
extraocular, bulbar, limb, and axial muscles (figure 2). cancer.34
60% of patients present with ptosis or diplopia, or AChR, MUSK, and LRP4 antibodies do not cross-react
both, and in 20% of patients, the disease is restricted with the heart muscle. In population studies,6 no
to ocular myasthenia gravis.1–4 Weakness of external eye increased mortality or morbidity related to cardiac
muscles is nearly always asymmetrical (figure 3), factors have been established. However, cardio-
whereas limb weakness is symmetrical and more physiological function can be marginally affected by
proximal than distal (figure 2).28 The variability in these antibodies.35 Many case reports of severe
symptoms in skeletal muscles is surprising because cardiomyositis and heart conduction abnormalities in
they all express the autoimmune target protein. This thymoma-associated myasthenia gravis and late-onset
variation results from many subtle factors affecting myasthenia gravis have been noted, most probably
neuromuscular transmission, muscle cell depolarisation induced by heart muscle autoimmunity. 36,37 Heart
or contraction, resistance to an immunological attack, function monitoring is recommended during severe
and regenerative capacity of myasthenia gravis exacerbations, especially in patients
muscle structures.2,17 with various antimuscle antibodies.38
A B C

Figure 3: Two patients with AChR-associated myasthenia gravis


Female patient with ophthalmoplegia (note adduction of right eye) and ptosis of the left eye (A). Male patient with opthalmoplegia (note the upward position of the
left eye) and ptosis of the right eye before treatment (B) and 1 year after immunosuppressive treatment (C).

Myasthenia gravis subgroups detectable AChR antibodies and


Early-onset myasthenia gravis with AChR antibodies generalised disease. About 30% of
Patients with early-onset myasthenia gravis have, by patients with a
definition, onset of their first symptom before age

50 years (table 1).1,7,39 Serum AChR antibodies are


detected by standard diagnostic testing. Patients with a
thymoma detected on imaging or during surgery are
excluded from this myasthenia gravis subgroup.
Thymic follicular hyperplasia occurs often but is not a
prerequisite, and this group responds to thymectomy.
Female cases outnumber male cases by three to one. 20,22
Early-onset myasthenia gravis has an association with
HLA-DR3, HLA-B8, and other autoimmune risk genes
(table 1),40,41 and all autoimmune disorders are more
widely reported in relatives of patients in this
myasthenia gravis subgroup.42 These findings suggest
subgroup differences in the pathogenesis of myasthenia
gravis.

Late-onset myasthenia gravis with AChR antibodies


Patients with late-onset myasthenia gravis are defined
as having their first onset of symptoms after age 50
years. In this group, serum AChR antibodies are
present, thymoma is not evident on imaging or during
surgery, and thymic hyperplasia occurs only rarely;
these patients most often will not respond to
thymectomy. The disease is slightly more frequently
reported in males than females, and weak HLA
associations occur with HLA- DR2, HLA-B7, and HLA-
DRB1*15:01.43

Thymoma-associated myasthenia gravis


Thymoma-associated myasthenia gravis is a para-
neoplastic disease. Myasthenia gravis is by far the most
widely reported autoimmune disease associated with a
thymoma, although pure red aplasia and
neuromyotonia are also associated with thymoma; this
association does not occur in other autoimmune
disorders. A thymoma is recorded in 10–15% of all
patients with myasthenia gravis. Nearly all have
thymoma develop myasthenia gravis, and even more have AChR
antibodies without myasthenia gravis. 44

MUSK-associated myasthenia gravis


MUSK is a protein expressed in the postsynaptic muscle membrane that is
functionally linked to AChR and necessary to maintain AChR function.
Overall, 1–4% of patients with myasthenia gravis have serum MUSK
antibodies, but more cases will probably be identified with increasingly
sensitive test assays. MUSK and AChR antibodies rarely coexist in the same
patient. MUSK- associated myasthenia gravis is usually reported in adults,
and rarely in the very old or in children. 45 No thymus pathological changes
are reported and patients usually have no response to thymectomy. IgG4
antibodies have an important role in the pathogenesis, and there is an HLA
association with HLA-DQ5,46–48 unlike in other myasthenia gravis
subgroups.
MUSK-associated myasthenia gravis shows pre- dominant involvement
of cranial and bulbar muscles. About a third of the patients present with
ptosis and diplopia.27 In more than 40% of patients with MUSK- associated
myasthenia gravis, bulbar weakness is a first symptom, with facial,
pharyngeal, and tongue weakness, often associated with neck and
respiratory involvement. Limb weakness is not common, and ocular
muscles are often unaffected.27 Little variation in muscle strength is
reported during the day, and muscle atrophy might occur.

LRP4-associated myasthenia gravis


LRP4 is expressed in the postsynaptic muscle membrane; it is a receptor
for nerve-derived agrin and an activator of MUSK, and is necessary to
maintain AChR function. LRP4 antibodies have been detected in 2–27% of
patients with myasthenia gravis without AChR and MUSK antibodies, with
a female pre- ponderance.49,50 Most of these patients present with ocular or
generalised mild myasthenia gravis, and about 20% of patients have
only ocular weakness for
more than 2 years. Respiratory insufficiency occurs
Myasthenia Age at onsetSex gravis HLA Thymus
very rarely, except in a subgroup with additional MUSK subgroup associations pathological
antibodies. In two-thirds of patients with LRP4- changes
Active immune response
associated myasthenia gravis, the thymus is atrophic
and normal for age, but hyperplasia has been reported. 5 AChREarly onset<50 years More female than male DR3-B8-A1Hyperplasia
Commercial tests are not yet available for LRP4
antibody testing, meaning that this group can be AChRLate onset>50 years More male than female Diverse Normal or
identified only by a few institutions. hyperplasia
AChR Thymoma Variabl ·· ·· Lymphoepitheliom
Antibody-negative generalised myasthenia gravis MUSK MUSK- e Substantially DR14, a Normal
Myasthenia gravis without detectable AChR, MUSK, or myastheni Variabl more female than DR16, DQ5
a gravis e male
LRP4 antibodies represents a heterogeneous group
LRP4 LRP4- Variabl ·· ·· Normal
pathogenically. Some patients have low-affinity anti- myastheni e
bodies or low concentration of antibodies to AChR, a gravis
MUSK, or LRP4 antigen targets, identified by cell-based Unknown SNMG Variabl ·· ·· Normal or
methods only, that are not detectable in routine e hyperplasi
assays.51,52 Low-affinity antibodies are pathogenic in Passive transfer of a
vivo, and the disease in patients with such antibodies is antibodies
AChR, or NeonatalNeonate Equal proportion ·· None
probably similar to that in the myasthenia gravis MUSK, or myasthenia of female to male
LEMSgravis
subgroup with detectable antibodies. Low-affinity
antibodies seem to account for 20–50% of patients in AChR=acetylcholine receptor. MUSK=muscle-specific kinase. LRP4=lipoprotein-related
the antibody-negative generalised myasthenia gravis protein 4.
SNMG=seronegative myasthenia gravis. LEMS=Lambert-Eaton myasthenic syndrome.
subgroup.51,52 Antibodies to agrin and cortactin often
occur in combination with other autoantibodies. 15,17,52 Table 1: Myasthenia gravis antibody and subgroup characteristics
Their functional relationship to other targeted proteins
is not clear. Some patients with myasthenia gravis elements of the thymus and are active in early-onset
probably have pathogenic antibodies against yet- myasthenia gravis, whereas thymoma cells contain
undefined antigens in the postsynaptic membrane. The muscle-specific antigens and have antigen-presenting
diagnosis is more challenging in patients in whom no properties.54 AChR expression can be activated in
specific autoantibodies are detected. In such patients, thymic epithelial cells through cytokine and receptor
non-myasthenia gravis myasthenic syndromes and signalling, potentially triggered by a virus; 3,55 however,
other muscle and non-muscle disorders should also be no specific virus has been identified so far. MicroRNAs
considered.3 can mediate immunoregulatory processes, be induced
by environ- mental events, and seem to be abnormally
Ocular myasthenia gravis expressed in myasthenia gravis.56 Autoreactive T cells,
In some patients with myasthenia gravis, the weakness specific for AChR, escape the normal intrathymic
is restricted to the ocular muscles. Patients with purely surveillance and are exported to the periphery where
ocular weakness are at risk of developing generalised they stimulate B cells to produce antibodies.
myasthenia gravis, especially early in the disease. 90% Differences in autoantibody pattern, HLA associations,
of those who have had the ocular form for more than 2 thymic pathological changes, cytokine intrathymic
years will remain in this subgroup. 53 Half of patients pattern, and T-cell subsets and clones all point to
with ocular myasthenia gravis have detectable AChR differences in induction mechanisms for early-onset,
antibodies, whereas MUSK antibodies very rarely late-onset, and thymoma-associated myasthenia
occur.53 gravis.44

Thymus pathological changes Neurophysiological testing


Thymoma, but no other thymic tumours, is associated Neurophysiological tests are unnecessary in patients
with myasthenia gravis. Thymic hyperplasia is reported with typical myasthenia gravis symptoms because
in most patients with early-onset myasthenia gravis diagnosis can be confirmed by specific antibody tests;
and in some patients with late-onset, ocular, and these tests are also not helpful for myasthenia gravis
antibody- negative disease. CT scanning or MRI of the subgroup classification. However, they are important
mediastinum should be undertaken in all patients with for correct diagnosis in patients with myasthenia gravis
myasthenia gravis to assess for a thymoma. 1–4,7 Both without detectable autoantibodies.
sensitivity and specificity are challenges for imaging. Repetitive nerve stimulation and single-fibre electro-
Experimental and clinical evidence strongly suggests myography for an increased jitter are useful tests for
that early-onset and thymoma-associated myasthenia patients with myasthenia gravis. Single-fibre testing is
gravis are initiated within the thymus.44 Myoid muscle- the most sensitive, whereas decrement at repetitive
like cells and professional antigen-presenting cells are stimulation is the most specific.1 Both sensitivity and
specificity rely on investigation quality. Even after patients for whom some doubt about diagnosis remains
combined neurophysiological and antibody testing, after testing, from our experience, do not have auto-
myasthenia gravis can be difficult to rule out. Most immune myasthenia gravis.

Drug Control or comparator Number of Duration Primary outcome ClinicalTrials. Result


participants measure gov number
Corticosteroids
Mount (1964)58 Corticotropin Placebo 43 12 weeks Eye movements ·· No significant difference
Howard et al (1976) 59 Alternate-day prednisone Placebo 13 24 weeks Clinical score ·· No significant difference
Lindberg et al (1998) 60 Pulse methylprednisone Placebo 19 2 weeks Muscle fatigue test ·· p<0·01
Benatar et al (2015)61 Prednisolone Placebo 11 16 weeks Treatment failure NCT00995722 Completed
Assistance Publique— Slow decrease of Rapid decrease 118 60 weeks Minimal NCT00987116 Ongoing
Hôpitaux de Paris prednisolone plus of prednisolone manifestation
(2009–2015) azathioprine plus
azathioprine
Azathioprine
Bromberg et al (1997) 62 Azathioprine Prednisone 10 52 weeks Observational ·· Descriptive
Palace et al (1998)63 Prednisolone and Prednisolone and placebo 34 156 weeks Prednisone dose ·· p=0·02
azathioprine
Ciclosporin
Tindall et al (1987)64 Ciclosporin Placebo 20 52 weeks QMGS, AChR titre ·· Only QMGS significant
Tindal et al (1993)65 Ciclosporin Placebo 39 26 weeks QMGS, AChR titre ·· p=0·004
Tacrolimus (FK506)
Nagane et al (2005)66 FK506 Placebo 34 52 weeks Prednisone dose ·· p<0·05
Yoshikawa et al (2011) 67 Tacrolimus Placebo 80 28 weeks Prednisone dose NCT00309088 No significant difference
Astellas Pharma Inc (2011– Tacrolimus Placebo 83 24 weeks QMGS NCT01325571 Ongoing
14)
Mycophenolate
Meriggioli et al (2003) 68 Mycophenolate mofetil Placebo 14 20 weeks QMGS ·· No significant difference,
except SFEMG (p=0.03)
Hoffmann-La Roche Mycophenolate mofetil Placebo 136 36 weeks Responder status NCT00683969 Completed
(2004–07)
Hoffmann-La Roche Mycophenolate mofetil Placebo 136 12–52 weeks Adverse events NCT00408213 Completed
(2004–07)
FDA Office of Orphan Mycophenolate mofetil Placebo 80 12 weeks QMGS NCT00285350 No significant difference
Products
Development/Duke
University, NC, USA (2008)
Sanders et al (2008)69 Mycophenolate mofetil Placebo 176 36 weeks Myasthenia gravis ·· No significant difference
composite
Qualitix Clinical Research Co Mycophenolate mofetil Azathioprine 40 52 weeks Remission NCT00997412 Completed
Ltd (2009–11)
Methotrexate
Pasnoor et al (2013) 70 Methotrexate Placebo 50 36 weeks Prednisone dose NCT00814138 Ongoing
Immunoglobulin or plasma exchange
Gajdos et al (1997) 71 Plasma exchange vs ·· 87 15 days Myasthenic muscular ·· No significant difference
intravenous score
immunoglobulin
Wolfe et al (2002) 72 Intravenous Placebo 15 6 weeks QMGS ·· No significant difference
immunoglobulin
Gajdos et al (2005) 73 Intravenous Placebo 173 2 weeks Myasthenic muscular ·· No significant difference
immunoglobulin score
(two doses)
Zinman et al (2007) 74 Intravenous Placebo 50 4 weeks QMGS NCT00306033 Intravenous
immunoglobulin immunoglobulin effective
(p<0.047)
Barth et al (2011)75 Intravenous Plasma exchange 84 2 weeks QMGS NCT01179893 Equally effective
immunoglobulin
Benesis Corporation Intravenous Plasma exchange 46 4 weeks QMGS NCT00515450 Completed
(2007–10) immunoglobulin
(GB-0998)
(Table 2 continues on next page)
Drug Control or comparator Number of Duration Primary outcome ClinicalTrials. Result
participants measure gov number
(Conintued from previous page)
Rituximab
Yale University, CT, USA Rituximab Placebo 50 52 weeks Prednisone dose NCT02110706 Ongoing
(2014–17) reduction
Eculizumab
Howard et al (2013)76 Eculizumab Placebo 14 18 weeks QMGS, adverse NCT00727194 p=0·0144
events
Alexion Pharmaceuticals Eculizumab Placebo 92 26 weeks Myasthenia gravis NCT01997229 Ongoing
(2013–16) ADL score
Belimumab
GlaxoSmithKline (2013–14) Belimumab Placebo 42 24 weeks QMGS NCT01480596 Ongoing
Leflunomide
First Affiliated Hospital, Sun Leflunomide Azathioprine 158 48 weeks Clinical response NCT01727193 Ongoing
Yat-Sen University, China
(2012–15)
Tirasemtiv (CK-2017357)
Sanders et al (2015) 77 CK-2017357 Placebo 32 2 days QMGS, VC, MMT NCT01268280 Completed

Thymectomy
University of Alabama at Thymectomy plus Prednisolone 150 3 years AU QMGS NCT00294658 Ongoing
Birmingham, AL, USA/ prednisolone
National Institute of
Neurological Disorders and
Stroke, USA (2008)

Treatment of myasthenia gravis effects have not been


Symptomatic drug treatment
Drugs that increase the amount of acetylcholine at
neuromuscular endplates after motor nerve stimulation
improve muscle weakness in all myasthenia gravis
subgroups; pyridostigmine is the preferred drug for
symptomatic treatment.7 Other acetylcholinesterase
inhibitors, such myasthenia
QMGS=quantitative as neostigmine and ambenonium
gravis score. AChR=acetylcholine receptor. SFEMG=Single-fibre electromyography. ADL=activities of daily living. VC=vital capacity. MMT=manual
muscle have
chloride, test. AU=area underdurations
different the curve. of action and can differ
regarding side-effects. The improvement reported in
Table 2: Randomised trials of treatments for autoimmune myasthenia gravis
patients with these drugs is so specific that it is used as a
diagnostic clue in patients who are antibody negative.
Reduction of acetylcholine breakdown by
acetylcholinesterase inhibition is the most effective
symptomatic treatment in myasthenia gravis, and is
better than increasing acetylcholine release
presynaptically, although a mild beneficial effect of
ephedrine or 3, 4-diaminopyridine might be seen. The
observational effects are so clear that randomised studies
have not been undertaken and are difficult to justify.57 In
MUSK-associated myasthenia gravis, acetylcholinesterase
inhibitors are less effective and induce frequent side-
effects.27 The optimum dose is a balance between
increased muscle strength and side-effects due to
cholinergic stimulation in the autonomic nervous system.
Glycopyrronium bromide, atropine sulfate, and
loperamide can be used to treat muscarinergic side-
effects. Long-term treatment with acetylcholinesterase
inhibitors is safe and habituation or cumulative side-
reported. Some patients with no
or only very mild symptoms
choose to continue to take an
acetylcholinesterase inhibitor.
This continuation might be out
of habit or concern of disease, or
because the inhibitors induce a
substantial subjective
improvement in these patients.

Immunosuppressive drug treatment


For patients with myasthenia
gravis in all subgroups who do
not have a fully satisfactory
functional result with
symptomatic treatment alone,
immunosuppressive drugs
should be initiated (tables 2,3,
and figure 4). Both treatment
effects and side-effects are
dose dependent. Finding the
optimum drug dose for each
patient is as important as
selecting the optimum drug. To
maximise effect and minimise
side-effects, a combination of
immunosuppressive drugs is
preferable for most patients.
Placebo-controlled studies and
those comparing alternative
treatments are rare.
Recommendations are
generally based on the sum of
many studies with weak
evidence, or on guidelines,
clinical experience, and
consensus reports.78 Formal
standards for patient
assessment can be helpful to
assess treatment response.79
Prednisone and prednisolone
improve muscle strength in all
myasthenia gravis subgroups.
Prednisone and prednisolone
are used in the same manner
and are equally effective.
Prednisone is activated by the
liver into prednisolone. The
beneficial effect manifests
after 2–6 weeks, faster than for
most other treatments. In a few
Number of Duration Study design ClinicalTrials.gov prednisolone should not be given as an alternate-day
Study period
participants number treatment to patients with diabetes because fluctuations
Bortezomib 18 6 months Open NCT02102594 in glucose concentrations result from this treatment
2014–16
GM-CSF 12 120 days Open NCT01555580 approach. If muscle strength differs for off-treatment
2012–13
2013–14 days and on-treatment days, a low dose (5–10 mg) of
Plasmapheresis 10 14 weeks Observational NCT01927692 prednisone or prednisolone can be added on off-days.
For
2004–09
Rituximab 10 12 months Open NCT00619671 ocular myasthenia gravis, observational studies53,82
2008–11
Rituximab 30 12 months Open NCT00774462 suggest that prednisolone treatment reduces the risk of
2002–16
Stem-cell therapy 10 5 years Open, phase 1 NCT00424489 developing generalised myasthenia gravis, although this
2014–17
Subcutaneous 25 12 weeks Open, phase 2 NCT02100969 observation has not been confirmed. For patients who
intravenous
immunoglobulin take long-term corticosteroids, specific precautions
Subcutaneous 10 6 months Open NCT01828294 2011–15 should be taken to reduce the risks of glucose
intravenous
immunoglobulin intolerance, gaining excess bodyweight, hypertension,
development of osteoporosis. A UK registry-based
Tacrolimus 11 28 weeks Open NCT00309101 2006–09 and 83
study did not report an increased fracture risk in
patients with
GM-CSF=granulocyte-macrophage colony stimulating factor.
myasthenia gravis.
Azathioprine is an effective drug for all myasthenia
Table 3: Ongoing non-randomised trials of treatments for autoimmune myasthenia gravis gravis subgroups, with 2–3 mg/kg being the most
registered in ClinicalTrials.gov

maximum effect. Prednisone or

Myasthenia gravis diagnosis confirmed

Start acetylcholine esterase inhibitor and


undertake thymectomy (if early onset or
thymoma)

Yes Continue with acetylcholine


Clinical esterase inhibitor
remission?
No

Start prednisolone and


azathioprine

Continue with prednisolone


Yes (lowest possible dose) and
Very good clinical
azathioprine
effect?
No

Start mycophenolate mofetil (mild,


moderate symptoms) or rituximab (severe
symptoms)

Yes Continue treatment


Sufficient
effect?

No immunosuppressive drugs
A Start other
(methotrexate, ciclosporin, tacrolimus)
B Re-evaluate myasthenia gravis

Figure 4: Treatment of generalised myasthenia gravis

patients, initial deterioration of generalised myasthenia


gravis has been reported lasting for up to 3 weeks.7,54,80
The starting dose is most often 0·75–1·0 mg/kg per
day1 for prednisone and prednisolone and is gradually
increased; alternate-day dosing is thought to reduce
side-effects and is recommended by some treatment
guidelines.2,3,81 After optimum improvement has been
induced, the drug dose should be gradually reduced,
and continued at the lowest dose necessary to obtain
www.thelancet.com/neurology Vol 14 October 1031
2015
This combination is often recommended as a first-choice treatment for
patients with generalised myasthenia gravis who need
immunosuppression, and is more beneficial than corticosteroids alone
with fewer side- effects. The azathioprine effect is delayed and from
clinical experience is usually seen after 6–15 months, and might further
increase during the subsequent 1–2 years.63 This makes the combination
with prednisolone convenient, and prednisolone can be reduced when the
azathioprine effect has been established. Regular follow- up is necessary
because of the risk of leucopenia and hepatotoxic effects, especially during
the first months of treatment. Low thiopurine methyltransferase activity
increases the risk for azathioprine toxic effects, and can be tested before
the start of treatment. Long-term treatment is also safe and effective in
young individuals.85 Azathioprine and corticosteroids in combination are
effective in almost all patients with myasthenia gravis. Patients with
ocular myasthenia gravis often respond well to a small dose (10–30 mg on
alternate days) of corticosteroids alone.
Mycophenolate mofetil is a prodrug that after conversion blocks purine
synthesis and interferes with B-cell and T-cell proliferation. Most
guidelines recommend the drug for mild and moderate myasthenia gravis
if the initial immunosuppressive therapy fails, 2–4 often together with
prednisolone. This recommendation is based on retrospective studies 1–3,8
and clinical experience. Mycophenolate mofetil is not recommended as
first-line treatment. In two prospective and controlled trials,69,86
mycophenolate mofetil did not show additional benefit when given as
initial treatment combined with prednisone. The studies had short
durations of only 12 weeks and 9 months. There were no stopping rules
for the use of corticosteroids and the lowest prednisone dose was 7·5 mg
per day, which might have obscured an effect of mycophenolate mofetil.
Little is known about myasthenia gravis subgroup responses for this
drug.87

www.thelancet.com/neurology Vol 14 October 1031


2015
insufficient
Side-effects are rare, with mild headache, nausea, and
diarrhoea the most commonly reported.
Rituximab has emerged as a potentially effective drug
in myasthenia gravis.81,88,89 It is a chimeric IgG1
monoclonal antibody that depletes all types of
B lymphocytes through specific binding to the
transmembrane CD20 antigen. This drug should, in our
opinion, be considered in moderate and especially
severe myasthenia gravis that does not respond
sufficiently to first-line immunosuppressive treatment.
However, controlled studies have not been done, and
rituximab is not regarded as a fully established
treatment. About two- thirds of patients with severe
myasthenia gravis and insufficient response to
prednisolone and azathioprine have a substantial
improvement on this treatment. 81,88–91 Open and
uncontrolled studies90–92 show that patients with MUSK-
associated myasthenia gravis in particular have a
favourable response, which is especially important as
this myasthenia gravis subgroup often has a lower
response to the first-line symptomatic and immuno-
suppressive treatment. In most reports, the induction
treatment recommended for rheumatological diseases
has been used, which is two doses of rituximab 1000
mg, and then another two doses of 1000 mg after 2
weeks.81,88–91 Lower doses have been suggested for
myasthenia gravis.88 Most centres would give additional
rituximab doses only to patients with deterioration
after a substantial and long- lasting response, and then
in the lowest effective dose.92 Rituximab is most often
combined with prednisolone and the combination with
prednisolone and azathioprine is also regarded as safe.
Severe side-effects have been reported as rare events
with rituximab for other autoimmune disorders,
including JC-virus-related pro- gressive multifocal
leukoencephalopathy, and have restricted the use of
rituximab in myasthenia gravis. Even in the absence of
controlled prospective studies and with high drug
costs, rituximab has, in our opinion, a place as an early
treatment for an increasing number of patients with
MUSK and AChR-associated myasthenia gravis.
Prospective and controlled studies have shown that
ciclosporin and methotrexate are effective as secondary
drugs for myasthenia gravis.65,70,93 The effect occurs in all
myasthenia gravis subgroups. Although comparative
studies have not been undertaken, ciclosporin and
methotrexate are thought to be as effective as
azathioprine.1–4,7 Patients should be monitored for
potential side-effects, especially nephrotoxic effects and
hypertension.
Tacrolimus has similarities to ciclosporin. A small
(34 patients) randomised but unblinded study 66 showed
that prednisone could be given at a reduced dose after
52 weeks when combined with tacrolimus. However, a
large double-blind study67 comprising of 80 patients did
not confirm this finding. The length of this study was
only 28 weeks and the therapeutic effect of prednisone
alone was better than expected.94 A new trial comparing
tacrolimus with placebo for patients with an
convincing data support surgery for this group.
response to glucocorticoids is in However, some guidelines7 recommend treating young
progress (NCT01325571). patients (up to age 60–65 years) with late-onset disease
Tacrolimus has an additional who have an enlarged thymus on imaging and no
effect on ryanodine receptor- antibodies to muscle titin or the ryanodine receptor,
mediated calcium release from similar to patients with early-onset myasthenia gravis.
the sarcoplasmic reticulum, For younger patients with late-onset myasthenia gravis,
which theoretically could lead to the thymus is most probably involved in the
improvements in muscle pathogenesis and the response to thymectomy would
strength in patients with be expected to be similar to that for early-onset disease.
myasthenia gravis. Thymectomy is not recommended for patients with
MUSK, LRP4, or ocular forms of myasthenia gravis as
Thymectomy no therapeutic effect has been shown. For patients with
Many studies have reported a generalised myasthenia gravis and low-affinity AChR
substantial effect of antibodies, thymus hyperplasia is usually impossible to
thymectomy in myasthenia establish by imaging. Such patients would be expected
gravis. These studies have to respond to thymectomy but cannot be distinguished
included control groups, but from other patients with myasthenia gravis who are
prospective and randomised found to be antibody negative.
studies have not been done. 1– Thymectomy should be done early, but is never an
3,7,95,96
For early-onset emergency; patients should be in a stable condition.
myasthenia gravis, we Intravenous immunoglobulin or plasma exchange
recommend a thymectomy immediately before surgery will improve the
early after symptom onset. All myasthenia gravis symptoms, reduce the risk of
thymus tissue needs to be complications, and contribute to a faster recovery.
removed. Video-assisted
thoracoscopic and robotic-
assisted methods are well
established, used by an
increasing number of centres,
and are usually preferred by
patients.97 Thymectomy can be
safe for juvenile myasthenia
gravis, down to an age of about
5 years.98 Improvement in
response to thymectomy occurs
gradually after some months,
and according to follow-up
studies, continues for up to 2
years postoperatively.95 No
other autoimmune disorders
have been shown to improve
after thymectomy.
Thymectomy should be
undertaken as an oncological
intervention when a thymoma
is detected or is strongly
suspected to avoid local
compression and spread to the
thoracic cavity. Any positive
effect on myasthenia gravis is
more unpredictable for the
thymoma than for the early-
onset subgroup.
Use of thymectomy in late-
onset myasthenia gravis is
debated. For patients with late-
onset disease with an atrophic
thymus or onset at age 60–65
years or older, thymectomy is
not recommended because no
Supportive treatment
Physical activity and low intensity and medium Treatment of myasthenia gravis crisis
intensity training provide short-term and long-term Crisis is defined as a need for intubation for respiratory
benefits for patients with myasthenia gravis. Weakness support caused by muscle weakness related to the
increases with repetitive muscle use, but patients with disease. Treatment includes intensive care with
myasthenia gravis can still find activities for which they respiratory support, treatment of infections, and
can adjust intensity and duration to increase their long- monitoring of vital functions and mobilisation (figure
term physical ability. Rest after such exercise is needed. 5). Intravenous immunoglobulin and plasma exchange
No controlled studies of myasthenia gravis training are specific immunosuppressive treatments with a
programmes have been published. rapid effect occurring after 2–5 days, and either one
Bodyweight control is important, as for other should be given to patients with severe myasthenia
disorders with muscle weakness. Such control is gravis exacerbations and always for crisis.99–103 These
especially relevant in patients with involvement of two treatment alternatives are equally effective, and
respiratory muscles. Infections in patients with can be given in sequence if necessary, as patients can
myasthenia gravis should be treated early and respond to one but not to the other. Standard protocols
vigorously because they can lead to myasthenia gravis include treatment for 3–6 consecutive days.
exacerbation and add to respiratory impairment. 1–4,7 Intravenous immunoglobulin is often slightly more
Drugs that interfere negatively with neuromuscular convenient and with a lower risk of severe side-effects,
transmission should be avoided. D-penicillamine and whereas plasma exchange might have a slightly faster
telithromycin should not be given to patients with effect. Catheter placement procedures for plasma
myasthenia gravis, and fluoroquinolones, exchange can be complex because access to large veins
aminoglycosides, macrolides, and neuromuscular is necessary. The treatment effect is usually restricted
blocking drugs will often cause worsening of the disease. to 2–3 months, owing to continuing antibody synthesis.
Neuromuscular blockade should be used with care during Plasma exchange and intravenous immunoglobulin can
anaesthesia. Sedatives that could suppress respiration be repeated when the effect tapers off. To secure long-
should be avoided in the treatment of patients with term improvement, this treatment is usually combined
severe myasthenia gravis. If a patient deteriorates when with standard immunosuppressive drugs, in higher
given a new drug, this drug should be withdrawn. doses than before the crisis or with add-on drugs. In
However, most patients with myasthenia gravis with patients with an acute exacerbation that does not
mild-to-moderate disease, or in stable remission, tolerate respond to intravenous immunoglobulin or plasma
drugs that have a relative warning, and most drugs can exchange, corticosteroids in high doses can be tried.
be used with caution. Myasthenia gravis crisis is a reversible condition.
Sometimes the treatment response is delayed, but
intensive care and vigorous immunosuppression should
Severe, generalised myasthenia be continued for as long as necessary, sometimes for
gravis several weeks.

Start intensive care, respirator if


necessary, start intravenous
Treatment of myasthenia gravis in pregnancy
immunoglobulin or plasma exchange Pregnancy does not affect myasthenia gravis in any
consistent way, with no increased risk of severe
Start or continue and intensify deterioration or myasthenia gravis crisis.85,104,105 During
Improvement
Yes long-term immunosuppressive the first weeks and few months post partum, the risk of
treatment
? symptom worsening is moderately increased, mainly
No because of stress and new demands.
Start plasma exchange or
Pyridostigmine and corticosteroids are regarded as
intravenous immunoglobulin safe treatments for pregnant women. 85 These drugs do
Start corticosteroids in high-dose not increase the risk of fetal malformations or delayed
methylprednisolone*
fetal development. Plasma exchange and intravenous
immunoglobulin can be used safely for exacerbations in
Yes
Continue and intensify pregnancy, and also as preparation for women giving
long-term immunosuppressive
Improvement
treatment birth. Evidence for potential teratogenic effects of other
?
immunosuppressive drugs is sparse. However, caution
No
is recommended for use of these drugs, and the
Start rituximab (outside intensive care manufacturers of immunosuppressive drugs generally
unit), continue intensive care and treat
complications advise against their use in pregnancy. Azathioprine has
been widely used for many years by young women with
AChR, MUSK, or LRP4 forms of myasthenia gravis. The
Figure 5: Treatment of severe myasthenia gravis exacerbations
general view is that this drug has very low, if any,
*1000 mg a day for 3 days.
increased teratogenic risk.85 Lactation should be
www.thelancet.com/neurology Vol 14 October
2015
10
encouraged in

www.thelancet.com/neurology Vol 14 October


2015
10
respond sufficiently to standard treatment, with a diagnosis confirmed by the
patients with myasthenia gravis, also for women on presence of
immunosuppressive drugs,105 but the passage of some
medications into breastmilk should be taken into
account. Mycophenolate mofetil and methotrexate have
teratogenic potential. Methotrexate might also reduce
female fertility. These two drugs should only rarely be
used in young women, and not in pregnancy.
Most female patients with myasthenia gravis give
birth in an uncomplicated way. Apart from the risk of
neonatal myasthenia gravis, no precautions are usually
needed. Caesarean section is not recommended as a
routine for these women, but should be considered in
prolonged births for women with moderate or severe
generalised myasthenia gravis because of muscle
fatigue.

Treatment of neonatal myasthenia gravis


Neonatal myasthenia gravis occurs in 10–15% of babies
of mothers with the disease. The cause of this transient
muscular weakness in these babies is transfer of the
mother’s AChR or MUSK antibodies of the IgG class
across the placenta. This weakness usually lasts for only
days or a few weeks and is typically mild but can
interfere with feeding and respiration. Mothers with
myasthenia gravis should always give birth at hospitals
experienced in respiratory support treatment for
newborn babies. The fact that neonatal myasthenia
gravis does not occur in all babies and that occurrence in
babies is not correlated with maternal disease severity
or AChR antibody concentration might be explained by
variation in AChR epitopes, epitope- binding affinity, and
non-AChR factors.105
Transplacental AChR antibodies can, in rare cases,
produce arthrogryposis due to severe intrauterine
movement inhibition. Such skeletal malformations were
reported in three of 127 babies in an unselected national
cohort.106 Arthrogryposis, AChR-antibody induced still-
births, and repeated spontaneous abortions can be
avoided by intravenous immunoglobulin infusions or
plasma exchange before and during pregnancy. This
treatment should be given in female patients with
myasthenia gravis who have already experienced such a
pregnancy outcome.

Conclusions and future directions


Most patients with myasthenia gravis do well and have
well controlled disease. However, most need long-term
and often life-long drug treatment with
acetylcholinesterase inhibitors and usually low-dose
immunosuppression. Pathogenic autoantibodies are
well characterised and myasthenia gravis subgroups
are defined accordingly. However, treatment is far from
antibody specific and is not even specific to the disease
subgroup. Many new and more traditional drugs that
have not been tested properly in myasthenia gravis
have modes of action that are expected to suppress
autoantibody production directly or indirectly, and
therefore might benefit patients with myasthenia
gravis. For patients with severe symptoms that do not
adapted treatment approach based on biomarker
(autoantibody) assessment and monitoring. The aim
Search strategy and selection criteria should be to suppress the anti- AChR, anti-MUSK, or
We searched MEDLINE and the Cochrane Library with the anti-LRP4 immune response without affecting other
terms “myasthenia gravis”, “myasthenic syndromes”, and immune reactions. An alternative approach could be
“myasthenia” from January 1995, to April, 2015. Guideline treatment that promotes tolerance to the antigens
and review papers were assessed in detail, and controlled (AChR, MUSK, and LRP4) that induce myasthenia
studies sought for in particular. Papers were selected by gravis.112 Patients with myasthenia gravis without
title and abstract. Only papers in English were included. detectable antibodies probably have pathogenic
Randomised trials on established and emerging therapies antibodies against undefined antigens in the
for myasthenia gravis are often scarce, so our neuromuscular junction; many proteins affect AChR
recommendations are based on the best available evidence or function, synthesis, and maintenance that could
clinical experience, where stated. potentially underlie antibody-negative disease. Auto-
immune myasthenia gravis with a T-cell-mediated and
autoantibodies and no non-antibody mechanism affecting neuromuscular
comorbidity as the symptom transmission could theoretically exist.
cause, such drugs could be When the causes of myasthenia gravis can be
tried, off-label, and with strict identified, they might be possible to avoid or prevent,
monitoring. These include potentially, for example, by vaccination. Until antigen-
monoclonal antibody drugs specific treatment is available, however, research
with a proven effect for other efforts should target new
autoimmune disorders.
Complement inhibition is one of
several potential strategies,76
with a focus on several factors
in the complement system.
Eculizumab, belimumab,
leflunomide, and etanercept are
drugs that might have the
potential to become new
myasthenia gravis treatment
options,76,107–109 although some
immunoactive drugs can
precipitate or worsen
myasthenia gravis.110 Tirasemtiv
(CK-2017357) selectively
sensitises fast skeletal muscle to
calcium by binding to its
troponin complex and amplifies
the muscle response when
neural input is diminished
secondary to neuromuscular
disease.111 A dose-related, short-
term improvement was
reported in a phase 2a
randomised placebo-controlled
trial.77 Any functionally relevant
long-term benefit to patients is
still to be proven. Several non-
antibody factors linked to the
immune system and skeletal
muscle affect the individual’s
muscle strength and immune
responses, and thereby each
patient’s myasthenia gravis
manifestations.
The high number of factors
associated with muscle
function in myasthenia gravis
should drive future research
towards an individually
18 Skeie GO, Mygland A, Treves S. Ryanodine
receptor antibodies in myasthenia gravis:
immunosuppressive drugs and drug combinations for epitope mapping and effect on calcium
the myasthenia gravis subgroups. Prospective and release in vitro. Muscle Nerve 2003; 27:
controlled studies should be encouraged and 81–89.
supported. Severe myasthenia gravis is a reversible 19 Romi F, Aarli JA, Gilhus NE. Myasthenia
gravis patients with ryanodine
disorder that should be treated with intensity and receptor antibodies have distinctive
optimism. clinical features. Eur J Neurol 2007;
14: 617–20.
Contributors
NEG planned the Review and wrote the first draft. JJV edited and
rewrote the first draft. Both authors searched primary sources for
information, produced tables and figures, and finalised the text.
Declaration of interests
NEG has received speaker’s honorarium from Octapharma, Baxter, and
Merck Serono. JJV is a partner in an FP7 European grant that is associated
with Curavac. The Department of Neurology at Leiden University Medical
Center has received fees from BioMarin for JJV’s consultancy work and
royalties from antibody tests. JJV received research grants from Prinses
Beatrix Spierfonds and National Institutes of Health.
References
1 Meriggioli MN, Sanders DB. Autoimmune myasthenia gravis:
emerging clinical and biological heterogeneity. Lancet Neurol 2009;
8: 475–99.
2 Gilhus NE. Myasthenia and neuromuscular junction.
Curr Opin Neurol 2012; 25: 523–29.

3 Querol L, Illa I. Myasthenia and the neuromuscular junction.


Curr Opin Neurol 2013: 26: 459–65.

4 Verschuuren JJ, Huijbers MG, Plomp JJ, et al. Pathophysiology of


myasthenia gravis with antibodies to the acetylcholine receptor,
muscle-specific kinase and low-density lipoprotein receptor-related
protein 4. Autoimmun Rev 2013; 12: 918–23.
5 Zisimopoulou P, Brenner T, Trakas N, Tzartos SJ. Serological
diagnostics in myasthenia gravis based on novel assays and recently
identified antigens. Autoimmun Rev 2013; 12: 924–30.
6 Owe JF, Daltveit AK, Gilhus NE. Causes of death among patients
with myasthenia gravis in Norway between 1951 and 2001.
J Neurol Neurosurg Psychiatry 2006; 77: 203–07.

7 Skeie GO, Apostolski S, Evoli A, et al. Guidelines for treatment of


autoimmune neuromuscular transmission disorders.
Eur J Neurol 2010; 17: 893–902.

8 Suh J, Goldstein JM, Nowak RJ. Clinical characteristics of refractory


myasthenia gravis patients. Yale J Biol Med 2013; 86: 255–60.
9 Jacob S, Viega S, Leite MI. Presence and pathogenic relevance of
antibodies to clustered acetylcholine receptor in ocular and
generalized myasthenia gravis. Arch Neurol 2012; 69: 994–1001.
10 Yang L, Maxwell S, Leite MI, et al. Non-radioactive serological
diagnosis of myasthenia gravis and clinical features of patients
from Tianjin, China. J Neurol Sci 2011; 301: 71–76.
11 Cole RN, Ghazanfari N, Ngo ST, et al. Patient autoantibodies deplete
postsynaptic muscle-specific kinase leading to dissembly of the ACh
receptor scaffold and myasthenia gravis in mice.
J Physiol 2010; 17: 3217–29.

12 Y, Ito M, Hirayama M, et al. Anti-MuSK autoantibodies block


binding of collagen Q to MuSK. Neurology 2011; 77: 1819–28.
13 Plomp JJ, Huijbers MG, Maarel SMVD, Verschuuren JJ. Pathogenic
IgG4 subclass autoantibodies in MuSK myasthenia gravis.
Ann NY Acad Sci 2012; 1275: 114–22.

14 Shen C, Lu Y, Zhang B, et al. Antibodies against low-density


lipoprotein receptor-related protein 4 induce myasthenia gravis.
J Clin Invest 2013; 123: 5190–202.
15 Gasperi C, Melms A, Schoser B, et al. Anti-agrin autoantibodies in
myasthenia gravis. Neurology 2014; 82: 1976–83.
16 Zhang B, Shen C, Bealmear B, et al. Autoantibodies to agrin in
myasthenia gravis. PLoS One 2014; 9: e91816.
17 Gallardo E, Martinez-Hernandez E, Titulaer MJ, et al. Cortactin
autoantibodies in myasthenia gravis. Autoimmun Rev 2014;
13: 1003–07.
20 Heldal AT, Owe JF, Gilhus NE, et al. Seropositive myasthenia gravis; a nationwide epidemiologic
study. Neurology 2009; 73: 150–51.
21 Andersen JB, Engeland A, Owe JF, et al. Myasthenia gravis requiring pyridostigmine treatment
in a national population cohort. Eur J Neurol 2010; 17: 1445–50.
22 Carr AS, Cardwell CR, McCarron PO, et al. A systematic review of population based
epidemiological studies in myasthenia gravis. BMC Neurol 2010; 10: 46
23 Pakzad Z, Aziz T, Oger J. Increasing incidence of myasthenia gravis among elderly in British
Columbia, Canada. Neurology 2011;
76: 1526–28.

24 Pedersen EG, Hallas J, Hansen K, et al. Late-onset myasthenia not on the increase: a
nationwide register study in Denmark, 1996–2009. Eur J Neurol 2012; 20: 942–48.
25 VanderPluym, J, Vajsar J, Jacob FD, et al. Clinical characteristics of pediatric myasthenia: a surveillance
study. Pediatrics 2013; 132: e939–44.
26 Niks EH, Kuks JBM, Verschuuren JJGM. Epidemiology of myasthenia gravis witht anti-muscle
specific kinaase antibodies in the Netherlands. J Neurol Neurosurg Psychiatry 2007; 78: 417–
18.
27 Guptill JT, Sanders DB, Evoli A. Anti-MuSK antibody myasthenia gravis; clinical findings and
response to treatment in two large cohorts. Muscle Nerve 2011; 44: 36–40.
28 Rodolico C, Toscano A, Autunno M, et al. Limb-girdle myasthenia; clinical, electrophysiological
and morphological features in familial and autoimmune cases. Neuromuscul Disord 2002; 12:
964–69.
29 Gilhus NE, Nacu A, Andersesn JB, Owe JF. Myasthenia gravis and risks for comorbidity. Eur J
Neurol 2015; 22: 17–23.
30 Liewluck T. Immune-mediated rippling muscle disease; another inflammatory myopathy in
myasthenia gravis. Arch Neurol 2010; 67: 896–97.
31 Mehanna R, Patton EL, Phan CL, Harati Y. Amyotrophic lateral sclerosis with positive anti-
acetylcholine receptor antibodies; case report and review of the literature. J Clin
Neuromuscul Dis 2012; 14: 82–85.
32 Filosso PL, Galassi C, Ruffini E, et al. Thymoma and the increased risk of developing
extrathymic malignancies: a multicentre study. Eur J Cardiothor Surg 2013; 44: 219–24.
33 Pedersen EG, Pottegard A, Hallas J, et al. Use of azathioprine for non-thymoma myasthenia
and risk of cancer: a nationwide case- control study in Denmark. Eur J Neurol 2013; 20:
942–48.
34 Pedersen EG, Pottegard A, Hallas J, et al. Risk of non-melanoma skin cancer in myasthenia
patients treated with azathioprine. Eur J Neurol 2014; 20: 942–48.
35 Owe JF, Davidsen ES, Eide GE, et al. Left ventricular long-axis function in myasthenia
gravis. J Neurol 2008; 255: 1777–84.
36 Suzuki S, Utsugisawa K, Yoshikawa H, et al. Autoimmune targets of heart and skeletal
muscles in myasthenia gravis. Arch Neurol 2009; 66: 1334–38.
37 Kumagai S, Kato T, Ozaki A, et al. Serial measurements of cardiac troponin I in patients with
myasthenia gravis-related cardiomyopathy. Int J Cardiol 2013; 168: e79–80.
38 Suzuki S, Utsugisawa K, Suzuki N. Overlooked non-motor symptoms in myasthenia gravis. J
Neurol Neurosurg Psychiatry 2013; 84: 989–94.
39 Alkhawajah NM, Oger J. Late-onset myasthenia gravis; a review when incidence in older adults
keeps increasing. Muscle Nerve 2013; 48: 705–10
40 Gregersen PK, Kosoy R, Lee AT, et al. Risk for myasthenia gravis maps to a (151) pro-ala
change in TNIP1 and human leukocyte antigen-B*08. Ann Neurol 2012; 72: 927–35.
41 Renton AE, Piner HA, Provenzano C, et al. A genome-wide association study of
myasthenia gravis. JAMA Neurol 2015; 72: 396–404.
42 Klein R, Marx A, Ströbel P, et al. Autoimmune associations and autoantibody screening show
focused recognition in patient subgroups with generalized myasthenia gravis. Hum Immunol
2013; 74: 1184–93.
43 Maniaol AH, Elsais A, Lorentzen ÅR, et al. Late onset myasthenia gravis is associated with HLA
DRB1*15:01 in the Norwegian population. PLoS One 2012; 7: e36603.
44 Marx A, Pfister P, Schalke B, et al. The different roles of the thymus in the pathogenesis of the
various myasthenia gravis subtypes. Autoimmun Rev 2013; 12: 875–84.
67 Yoshikawa H, Kiuchi T, Saida T, et al. Randomised, double-blind, placebo-controlled study of
tacrolimus in myasthenia gravis.
45 Skjei KL, Lennon VA, Kuntz NL. Muscle specific kinase J Neurol Neurosurg Psychiatry 2011; 82: 970–77.
autoimmune myasthenia gravis in children: a case series.
Neuromuscul Disord 2013; 23: 874–82.
46 Bartoccioni E, Scuderi F, Augugiaro A, et al. HLA class II allele
analysis in MuSK-positive myasthenia gravis suggests a role for
DQ5. Neurology 2009; 72: 195–97.
47 Klooster R, Plomp JJ, Huijbers MG, et al. Muscle-specific kinase
myasthenia gravis IgG4 autoantibodies cause severe
neuromuscular junction dysfunction in mice. Brain 2012;
135: 1081–101.

48 Alahgholi-Hajibehzad M, Yilmaz V, Guisen-Parman Y, et al.


Association of HLA-DR131*14,-DR131*16 and –DQB1*05 with
MuSK-myasthenia gravis in patients from Turkey. Hum Immunol
2013; 74: 1633–35.
49 Higuchi O, Hamuo J, Motomura M, et al. Autoantibodies to low-
density lipoprotein receptor-related protein 4 in myasthenia gravis.
Ann Neurol 2011; 69: 418–22.
50 Zhang B, Tzartos JS, Viegas S, et al. Autoantibodies to
lipoprotein-related protein 4 in patients with double-negative
myasthenia gravis. Arch Neurol 2012; 69: 445–51
51 Leite MI, Jacob S, Viegas S, et al. IgG1 antibodies to acetylcholine
receptors in ”seronegative” myasthenia gravis. Brain 2008;
131: 1940–52.

52 Cossins J, Belaya K, Zoltowska K, et al. The search for new


antigenic targets in myasthenia gravis. Ann N Y Acad Sci 2013;
1275: 123–28.
53 Kerty E, Elsais A, Argov Z, Evoli A, Gilhus NE. EFNS/ENS
guidelines for the treatment of ocular myasthenia gravis.
Eur J Neurol; 2014: 21: 687–93.
54 Romi F, Bo L, Skeie GO, et al. Titin and ryanodine receptor epitopes
are expressed in cortical thymoma along with costimulatory
molecules. J Neuroimmunol 2002; 128: 82–89.
55 Cufi P, Dragin N, Weiss JM, et al. Implication of double-stranded
RNA signaling in the etiology of autoimmune myasthenia gravis.
Ann Neurol 2012; 73: 281–93.
56 Panse RL, Berrih-Aknin S. Autoimmune myasthenia gravis:
autoantibody mechanisms and new developments on immune
regulation. Curr Opin Neurol 2013; 26: 569–76.
57 Mehndiratta MM, Pandey S, Kuntzer T. Acetylcholinesterase
inhibitor treatment for myasthenia gravis.
Cochrane Database Syst Rev 2014; CD006986.

58 Mount, FW. Corticotropin in treatment of ocular myasthenia; a


controlled clinical trial. Arch Neurol 1964; 11: 114–24.
59 Howard FM Jr, Duane DD, Lambert EH, Daube JR. Alternate-day
prednisone: preliminary report of a double-blind controlled study.
Ann N Y Acad Sci 1976; 274: 596–607.
60 Lindberg C1, Andersen O, Lefvert AK. Treatment of myasthenia
gravis with methylprednisolone pulse: a double blind study. Acta
Neurol Scand 1998; 97: 370–73.
61 Benatar M, McDermott MP, Sanders DB, et al, for the Muscle Study
Group (MSG). Efficacy of Prednisone for the Treatment of Ocular
Myasthenia (EPITOME): a randomized controlled trial. Muscle
Nerve 2015; published online July 14. DOI:10.1002/ mus.24769.
62 Bromberg MB, Wald JJ, Forshew DA, et al. Randomized trial of
azathioprine or prednisone for initial immunosuppressive
treatment of myasthenia gravis. J Neurol Sci 1997; 150: 59–62.
63 Palace J, Newsom-Davis J, Lecky B. A randomized double-blind trial of
prednisolone alone or with azathioprine in myasthenia gravis.
Neurology 1998; 50: 1778–83.
64 Tindall RS, Rollins JA, Phillips JT, Greenlee RG, Wells L, Belendiuk G.
Preliminary results of a double-blind, randomized, placebo-
controlled trial of cyclosporine in myasthenia gravis.
N Engl J Med 1987; 316: 719–24.

65 Tindall RS, Philips JT, Rollins JA, et al. A clinical therapeutic trial of
cyclosporine in myasthenia gravis. Ann N Y Acad Sci 1993;
681: 539–51.

66 Nagane Y, Utsugisawa K, Obara D, et al. Efficacy of low-dose FK506 in


the treatment of myasthenia gravis; a randomized pilot study. Eur
Neurol 2005; 53: 146–50.
the efficacy of prednisone in the treatment of ocular myasthenia
(EPITOME) trial. Ann N Y Acad Sci 2012; 1275: 17–22.
68 Meriggioli MN, Rowin J, Richman JG,
81 Benveniste O, Hilton-Jones D. The role of rituximab in the
Leurgans S. Mycophenolate mofetil
treatment of myasthenia gravis. Eur Neurol Rev 2010; 5: 95–
for myasthenia gravis: a double-
100.
blind, placebo-controlled pilot
study. Ann N Y Acad Sci 2003; 998: 82 Benatar M, Kaminski H. Medical and surgical treatment for ocular
494–9. myasthenia. Cochrane Database Syst Rev 2012; CD005081.
69 Sanders DB, Hart IK, Mantegazza R, 83 Pouwels S, Boer AD, Javaid MK, et al. Fracture rates in patients with
et al. An international, phase III, myasthenia gravis; the general practice research database.
randomized trial of mycophenolate Osteoporos Int 2013; 24: 467–76.
mofetil in myasthenia gravis. 84 Marinkovic G, Kroon J, Hoogenboezem M, et al. Inhibition of
Neurology 2008; 71: 400–06. GTPase rac1 in endothelium by 6-mercaptpurine results in
70 Pasnoor M, He J, Herbelin L, et al. immunosuppression in nonimmune cells; new target for an old
Phase II trial of methotrexate in drug. J Immunol 2014; 192: 4370–78.
myasthenia gravis. Ann N Y Acad 85 Norwood F, Dhanjal M, Hill M, et al. Myasthenia in pregnancy; best
Sci 2013; 1275: 23–28. practice guidelines from a UK multispeciality working group. J
71 Gajdos P, Chevret S, Clair B, Neurol Neurosurg Psychiatry 2014; 85: 538–43.
Tranchant C, Chastang C. Clinical trial 86 The Muscle Study Group. A trial of mycophenolate mofetil with
of plasma exchange and high-dose prednisone as initial immunotherapy in myasthenia gravis.
intravenous immunoglobulin in Neurology 2008; 71: 394–399.
myasthenia gravis. Myasthenia Gravis 87 Hehir MK, Burns TM, Alpers J, et al. Mycophenolate mofetil in
Clinical Study Group. AChR-antibody-positive myasthenia gravis; outcomes in
Ann Neurol 1997; 41: 789–96. 102 patients. Muscle Nerve 2010; 41: 593–98.

72 Wolfe GI, Barohn RJ, Foster BM, et 88 Blum S, Gillis D, Brown H, et al. Use and monitoring of low dose
al, for the Myasthenia Gravis-IVIG rituximab in myasthenia gravis. J Neurol Neurosurg Psychiatry
Study Group. Randomized, 2011; 82: 659–63.
controlled trial of intravenous 89 Maddison P, McConville J, Farrugia ME, et al. The use of rituximab in
immunoglobulin in myasthenia myasthenia gravis and Lambert-Eaton myasthenic syndrome.
gravis. Muscle Nerve 2002; 26: J Neurol Neurosurg Psychiatry 2011; 82: 671–73.
549–52.
73 Gajdos P, Tranchant C, Clair B, et al, 90 Diaz-Manera J, Martinez-Hernandez E, Querol L, et al. Long-
for the Myasthenia Gravis Clinical lasting treatment of rituximab in MuSK myasthenia.
Study Group. Treatment of Neurology 2012; 78: 189–93.
myasthenia gravis exacerbation with
intravenous immunoglobulin: a
randomized double-blind clinical
trial. Arch Neurol 2005; 62: 1689–
93.
74 Zinman L, Ng E, Bril V. IV
immunoglobulin in patients with
myasthenia gravis: a randomized
controlled trial. Neurology 2007;
68: 837–41.
75 Barth D, Nabavi Nouri M, Ng E, Nwe
P, Bril V. Comparison of IVIg and PLEX
in patients with myasthenia gravis.
Neurology 2011;
76: 2017-23.

76 Howard JF, Barohn PJ, Cutter GR, et al. A randomized,


double-blind, placebo-controlled
phase II study of eculizumab in
patients with refractory generalized
myasthenia gravis. Muscle Nerve
2013; 48: 76–84.

77 Sanders DB, Rosenfeld J, Dimachkie


MM, et al. A double-blinded,
randomized, placebo-controlled trial
to evaluate efficacy, safety and
tolerability of single doses of tirasemtiv
in patients with acetylcholine receptor-
binding antibody-positive myasthenia
gravis. Neurotherapeutics 2015; 12:
455–60.
78 Hart IK, Sthasiam S, Sharshar T.
Immunosuppressive agents for
myasthenia gravis (review).
Cochrane Database Syst Rev 2007;
CD005224.
79 Jaretzki III A, Barohn RJ, Ernstoff RM
et al. Myasthenia gravis:
recommendations for clinical
research standards. Ann Thorac
Surg 2000; 70: 327–34.
80 Benatar M, Sanders DB, Wolfe GI,
McDermott MP, Tawil R. Design of
91 Keung B, Robeson KR, DiCapua DB, et al. Long-term benefit of rituximab in MuSK autoantibody myasthenia gravis patients.
J Neurol Neurosurg Psychiatry 2013; 84: 1407–09.

92 Yi JS, DeCroos EC, Sanders DB et al. Prolonged B-cell depletion in MuSK myasthenia gravis following rituximab treatment.
Muscle Nerve 2013; 48: 992–93.

93 Heckmann JM, Rawoot A, Bateman K, et al. A single-blinded trial of methotrexate versus azathioprine as steroid-sparing agents in generalized
myasthenia gravis. BMC Neurol 2011; 11: 97.
94 Benatar M, Sanders D. The importance of studying history; lessons learnt from a trial of tacrolimus in myasthenia gravis.
J Neurol Neurosurg Psychiatry 2011; 82: 945.

95 Gronseth GH, Barohn RJ. Thymectomy for autoimmune myasthenia gravis (an evidence-based review). Neurology 2000; 55: 7–15.
96 Cea G, Benatar M, Verdugo RJ, Salinas RA. Thymectomy for non- thymomatous myasthenia gravis. Cochrane Database Syst Rev 2013; cd008111.
97 Ye B, Tantal J-C, Li W, et al. Video-assisted thoracoscopic surgery versus robotic-assisted thoracoscopic surgery in the surgical treatment of Masaoka
stage I thymoma. World J Surg Oncol 2013; 11: 157–62.
98 Liew WKM, Kang PB. Update on juvenile myasthenia gravis.
Curr Opin Pediatr 2013; 25: 694–700.

99 Gajdos P, Chevret S, Toyka KV. Plasma exchange for generalised myasthenia gravis. Cochrane Database Syst Rev 2002; CD002275.
100 Mandawat A, Kaminski H, Cutter G, et al. Comparative analysis of therapeutic options used for myasthenia gravis. Ann Neurol 2010; 68: 797–805.
101 Barth D, Nabavi N, Ng E, et al. Comparison of IVIg and PLEX in patients with myasthenia gravis. Neurology 2011; 76: 2017–23.
102 Gilhus NE. Acute treatment for myasthenia gravis. Nat Rev Neurol
2011; 7: 132–34.
103 Gajdos P, Chevret S, Toyka KV. Intravenous immunoglobulin for myasthenia gravis. Cochrane Database Syst Rev 2012; CD002277.
104 Ferrero S, Pretta S, Nicoletti A, et al. Myasthenia gravis: management issues during pregnancy.
Eur J Obst Gynecol Reproduct Biol 2005; 121: 129–38.

105 Hoff JM, Daltveit AK, Gilhus NE. Myasthenia gravis in pregnancy and birth: identifying risk factors, optimising care. Eur J Neurol 2007; 14: 38–43.
106 Hoff JM, Loane M, Gilhus NE, et al. Arthrogryposis multiplex congenital: an epidemiologic study of nearly 9 million births in 24 Eurocat registers. Eur J Obst Gynecol
Reproduct Biol 2011; 159: 347–50.
107 Ragheb S, Lisak R, Lewis R, et al. A potential role for B-cell activating factor in the pathogenesis of autoimmune myasthenia gravis. Arch Neurol 2008; 65: 1358–62.
108 Gomez AM, Vrolix K, Martinez-Martinez P, et al. Proteasome inhibition with bortezomib depletes plasma cells and autoantibodies in experimental autoimmune myasthenia
gravis. J Immunol 2011; 186: 2503–13.
109 Rowin J, Thiruppathi M, Arrebamen E, et al. Granulocyte macrophage colony-stimulating factor treatment of a patient in myasthenic crisis; effects on regulatory T cells. Muscle
Nerve 2012; 46: 449–53.
110 Fee DB, Kasarskis EJ. Myasthenia gravis associated with etanercept therapy. Muscle Nerve 2009; 39: 866–70.
111 Russell AJ, Hartman JJ, Hinken AC, et al. Activation of fast skeletal muscle troponin as a potential therapeutic approach for treating neuromuscular diseases. Nat Med 2012; 18:
452–56.
112 Steinman L. The road not taken: antigen-specific therapy and neuroinflammatory disease. JAMA Neurol 2013; 70: 1100–01.
Pediatric Myasthenia Gravis
Jason H. Peragallo, MD*,†

Myasthenia gravis is a disorder of neuromuscular transmission that leads to fatigue of skeletal


muscles and fluctuating weakness. Myasthenia that affects children can be classified into the
following 3 forms: transient neonatal myasthenia, congenital myasthenic syndromes, and juvenile
myasthenia gravis (JMG). JMG is an autoimmune disorder that has a tendency to affect the
extraocular muscles, but can also affect all skeletal muscles leading to generalized weakness and
fatigability. Respiratory muscles may be involved leading to respiratory failure requiring ventilator
support. Diagnosis should be suspected clinically, and confirmatory diagnostic testing be
performed, including serum acetylcholine receptor antibodies, repetitive nerve stimulation, and
electromyography. Treatment for JMG includes acetylcholinesterase inhibitors, immunosuppressive
medications, plasma exchange, intravenous immunoglobulins, and thymectomy. Children with
myasthenia gravis require monitoring by a pediatric ophthalmologist for the development of
amblyopia from ptosis or strabismus.
Semin Pediatr Neurol 24:116-121 C 2017 Elsevier Inc. All rights reserved.

Introduction
The myasthenic syndromes are due to defects of transmission
Pathophysiology
at the neuromuscular junction. Myasthenic syndromes in In normal synaptic transmission in the neuromuscular junc- tion,
children have 3 distinct forms with pathophysiologically the axon is depolarized and this depolarization travels to the axon
different mechanisms. The congenital myasthenic syndromes terminal. Voltage-gated calcium channels open, leading to
(CMS) are a group of genetic disorders that lead to muscle acetylcholine containing vesicles to fuse to the cell membrane.
weakness through structural or functional abnormalities of the Acetylcholine is then released from synaptic vesicles into the
proteins involved in neuromuscular transmission itself. synaptic cleft from the axon terminal. The acetylcholine travels
Transplacental transfer of maternal antibodies of a myasthenic across the synaptic cleft to the AChR sites where binding causes
mother causes transient neonatal myasthenia (TNM) in infants. sodium channels to open, depolarizing the motor end plate of the
Juvenile myasthenia gravis (JMG) is an autoimmune disorder muscle fiber. Acetylcholinesterase degrades the acetylcholine, and
that leads to dysfunction of acetylcholine receptors (AChR). All the sodium channels close, allowing repolarization of the muscle
forms of myasthenia lead to muscle fatigue and weakness of fiber.
varying degrees. Fluctuations in weakness are a hallmark of
this disease. If weakness involves the musculature involved in
respiration, these diseases can be life threatening.1,2 This
review will focus primarily on the JMG form. Congenital Myasthenic Syndromes
The CMS are not autoimmune diseases, but are a group of
disorders caused by alterations in the structure or function of
proteins in the neuromuscular junction. These disorders must
be suspected to distinguish them from myopathies or other
From the *Department of Ophthalmology, Emory University School of neurogenic conditions.3 The disease can be the result of, for
Medicine, Atlanta, GA. example, inability to form normal acetylcholine, inability to

Department of Pediatrics, Emory University School of Medicine, Atlanta, GA. transport and release acetylcholine, abnormal nicotinic AChR,
primary AChR decifiency, end plate development and main-
Supported in part by an unrestricted departmental Grant (Department of tenance protein abnormalities, defects in protein glycosylation,
Ophthalmology) from Research to Prevent Blindness, Inc, New York, and many others.4 The most common cause of CMS is
and by NIH/NEI core Grant P30-EY06360 (Department of alterations in the structure, presence, or kinetics of the AChR
Ophthalmology). itself.4 Performing a genetic analysis for the specific mutation
Address reprint requests to Jason H. Peragallo, MD, Department of Ophthal- causing CMS is necessary to direct treatment for that specific
mology, Emory University School of Medicine, 1365B Clifton Rd NE, Atlanta,
GA 30322. E-mail: jperaga@emory.edu

116 http://dx.doi.org/10.1016/j.spen.2017.04.003
1071-9091/11/& 2017 Elsevier Inc. All rights reserved.
Pediatric myasthenia gravis 11
7
syndrome. A treatment modality for CMS caused by one
mutation may actually be harmful to the patient if a B-cell–mediated disease, recent studies have proposed B-cell–
different mutation is present, and treatment must be activating factor as playing a role in its pathogenesis and as a
tailored to the specific mutation causing CMS. Children marker of disease activity in JMG.15,16
with CMS often will have a relative who is similarly
affected. They will present with fatigable weakness, usually
at a very early age, and can present in infancy. The affected Demographics
muscles may be ocular alone, which can lead to isolated
fatigable ptosis, or all skeletal muscles may be affected There have been varying estimates of the incidence of
leading to more widespread weakness. Investiga- tions for myasthenia gravis occurring in the general population in
autoantibodies found in JMG and TNM will be negative. the literature ranging from 1.7-30.0 cases per million
Making the distinction between CMS and JMG may be person years, with a prevalence of 77.7 cases per million
difficult if a known genetic mutation causative for CMS persons.17,18 Many studies estimating the incidence and
cannot be identified and autoantibodies leading to JMG are prevalence of JMG have also been published. A meta-
not detected. analysis of the literature on this subject concluded that the
incidence of JMG is approximately between 1.0 and 5.0
cases per million person years.17 Pediatric patients were
Transient Neonatal Myasthenia found to make up approximately 10%-15% of all patients
with autoimmune myasthenia gravis in a prevalence study
TNM is an antibody-mediated disorder that differs from in Virginia.19 Among Asian populations, there appears to be
JMG and CMS. This disease presents shortly after birth in a higher prevalence of JMG compared to white populations,
infants of mothers who have autoimmune myasthenia with JMG representing up to 50% of all autoimmune
gravis. The circulating maternal autoantibodies cross the myasthenia gravis cases.20,21
placenta to the fetus, leading to an affected child after birth.
Children of mothers who have myasthenia gravis will
develop TNM approximately 5%-30% of the time.5,6 Infants Clinical Presentation
can present with generalized hypotonia, feeble cry,
respiratory distress, poor suck, and extraocular muscle Fatigable skeletal muscle weakness that fluctuates is the
weakness.6 Symptoms are usually self-limited but characteristic sign of myasthenia gravis. Myasthenia gravis is
respiratory support may be necessary. Patients respond known to preferentially target the extraocular muscles. Cases
well to neostigmine treatment and plasma exchange. TNM in which only the extraocular muscles are affected are termed
can be distinguished from CMS by the detection of ocular myasthenia gravis (OMG). If any other skeletal muscles
autoantibodies, and when the disease is demon- strated to are involved, the patient is diagnosed with generalized
be transient once autoantibodies are cleared from the myasthenia gravis. The vast majority of patients who have
infant and symptoms resolve. No long-term treatment is myasthenia gravis will have involvement of the extraocular
required once the autoantibodies are no longer present. muscles at some point during the course of their disease.

Juvenile Myasthenia Gravis Ocular Myasthenia Gravis


Juvenile myasthenia gravis is an autoimmune disorder pre- Approximately 10%-35% of all cases of JMG are of the
senting with fatigability before the age of 19 years, which in OMG subtype.1,22 OMG is a more common form of JMG in
contrast to the CMS is not due to a structural abnormality in Asian populations than in other populations.20,21 The
the neuromuscular junction.7 JMG is also not a transient muscles involved include the vertical and horizontal rectus
disease, such as TNM. Autoantibodies against targets such muscles, inferior and superior oblique muscles, orbicularis
as the AChR and muscle-specific kinase (MuSK) lead to oculi, and the levator palperae superioris. The initial
decreased AChR activity though complement-mediated presentation in these children may be ptosis, strabismus, or
lysis of the postsynaptic membrane, increased rate of diplopia. In one community-based case-control study, JMG
degradation of the receptor, and direct inhibition of was the underlying cause of ptosis in 0.81% of pediatric
activity.8-10 Lipoprotein receptor–related membrane patients.23 JMG was present in children with ptosis at 26.7-
protein 4 (LRP-4) is another target of autoantibodies fold the rate of children who did not have ptosis.23 Patients
implicated in the development of myasthenia gravis in may actually have eyelid retraction, most prominent upon
patients in whom the antibodies to the AChR or MuSK were awakening, of the eyelid that is found to be ptotic in the
not detected.11 LRP-4 is a protein necessary for MuSK clinic. Directed questions about ptosis should address not
activation.12 Up to 18.4% of patients who do not have only if it is worse in the evenings, but also if it appears
antibodies to the AChR or MuSK were found to have resolved upon awakening in the morning.
antibodies to LRP-4 in one analysis.12 Other autoimmune Patients with JMG with or without the purely ocular
diseases, such as Hashimoto disease and autoimmune form of disease can present with pseudo-cranial nerve
polymyositis, may be present in the same individual who palsies, and may have variable strabismus measurements
has JMG.13 A recent study analyzed data from patients who when presenting to a pediatric ophthalmologist for
had both myasthenia gravis and aquaporin 4 antibody– evaluation for strabismus sur- gery. It is important to note
positive neuromyelitis optica.14 In this study, 20% of that the pupils are not involved to a clinically detectable
patients had JMG before developing neuromyelitis optica.14 degree in myasthenia gravis, distinguishing
As JMG is a
this disease from other neurologic causes of strabismus.
Pupillary abnormalities should prompt evaluation for other Having the patient look in downgaze causes relative rest of
neurological causes. Rates of progression from OMG to the levator palpebrae, which is “overstimulated” upon
generalized myasthenia gravis has been reported from 8%- upgaze.
49%, with rates of conversion for JMG specifically ranging Another simple in office test that can suggest myasthenia
from 8%-33%.2,24-29 Preadolescent patients with OMG may gravis if positive is the ice pack test. After measuring the
be less likely to progress to the generalized form than initial palperbral fissure height, an ice-filled glove or ice
adolescents.25 pack is placed over the patient’s eye or eyes. The ice is left
there for 1-2 minutes while the patient rests. Once the ice is
removed, the palpebral fissure is measured again. An
improvement of ptosis of 2 mm of more is a positive result.
Generalized Myasthenia Gravis This test has been found to be approximately 80%
The generalized form of JMG can be life threatening, as any sensitive for the detection of myasthe- nia gravis.31,32 An
skeletal muscle can be involved, including the muscles alternative to the ice pack test is the rest test, in which a
involved in respiration. If this occurs, patients may develop patient is asked to rest for 30 minutes after being
respiratory failure requiring intubation.1,8 Presenting signs can examined. Following a period of rest, the examination is
include the ocular motility abnormalities and ptosis described repeated, and measurements compared.33 The ice pack test
earlier, but must include other limb or facial weakness, may be more sensitive than the rest test for the detection of
difficulty breathing or swallowing, and chocking. Speech can myasthenia gravis.34
be affected leading to a change in voice characteristics.
Severity of myasthenia gravis can be classified according to the
Myasthenia Gravis Foundation of America’s classification
system, which allows for uniform criteria to be used cross- Serologic Testing
institution for research purposes and for monitoring disease Acetylcholine receptor antibody testing is readily available
severity.30 commercially. The 3 main AChR antibodies tested are
binding, blocking, and modulating antibodies. Their
presence is important as it can definitively make the
diagnosis of myas- thenia gravis in uncertain cases.8,35 One
Diagnosis study of patients with JMG found an AChR antibody
positivity rate of 72% of those presenting with generalized
Making the diagnosis of myasthenia gravis begins when a symptoms, and 41% in those presenting with ocular
patient demonstrates some form of fatigable weakness. A symptoms alone.36 Patients who were initially AChR
thorough targeted clinical examination should include some antibody negative became positive 41% of the time within
evaluation of fatigability to address suspicion of myasthenia 2 years.36 However, most of those patients who converted
gravis. Ancillary testing for its diagnosis includes serologic, to positive AChR antibody status did not have worsening of
pharmacologic, and electrophysiologic testing. symptoms. Remission was associated with conversion from
AChR antibody positive to AChR antibody–negative status.36
Another more recent study found an AChR antibody
Clinical Examination Techniques positivity rate of 86% in patients with JMG.22 The finding
Fatigability on sustained upgaze is one of the simplest tests that patients with the ocular form have autoantibodies to
to perform on patients in whom JMG is suspected. First, the AchR less frequently than the generalized form is
palpebral fissure height is taken and an extraocular similar to what is found in adults.37 Patients who are
motility examination is performed. The patient is then negative for AChR antibodies can also be tested for anti-
directed to look at a target above their heads, and MuSK antibodies, which have been found to be positive in
instructed to look at the target without moving their head up to 40% of all patients with autoimmune myasthenia
out of primary position. The target is held there for a gravis who are AChR antibody negative.38 Similar rates
certain period while the eyelid and ocular alignment is have been found in AChR antibody–negative JMG series.39
closely observed. Patients with JMG may demonstrate a Children who have anti-MuSK antibodies tend to have more
change in palpebral height and ocular alignment while severe, acute, generalized symptoms, with respiratory
being observed, which suggests fatigability. The target can distress, similar to what is found in adults with anti-MuSK
then be removed, and the ocular motility examination and antibodies.39,40 Response to treatment for JMG due to anti-
measurement of palpebral fissure height can be repeated. MuSK antibodies can be variable.39 Patients who are
Changes in these measurements suggest the presence of negative for both AChR antibodies and MuSK antibodies but
myasthenia gravis. clinically have myasthe- nia gravis are called “double
The Cogan lid twitch is another feature that can be seen seronegative”. Another antibody that has been found to be
in patients who have levator palpebrae superioris positive in double-seronegative patients is the lipoprotein
involvement leading to ptosis. This sign can be elicited by receptor-related membrane protein 4 (LRP-4).41 Pediatric
having a patient look in downgaze for a certain period, patients may be affected in anti-LRP- 4–mediated
followed by asking them to quickly look up. The eyelid myasthenia gravis as well.12 The incidence of anti-LRP-4
overelevates briefly (this is the “twitch”), followed by positivity varies widely from study to study, likely due to
fatigue and return to a ptotic position. different assays used by research teams.42 Anti-LRP-4–
associated myasthenia gravis tends to manifest mild
symptoms.12,41,42
Pharmacologic Testing
Immunosuppressive Agents
The classic pharmacologic test for myasthenia gravis uses a
short-acting antiacetylcholinesterase, edrophonium, in the The most common immunosuppressive agent prescribed
edrophonium (Tensilon) test. By inhibiting acetylcholinester- for myasthenia gravis is prednisone. This medication is
ase, the amount of acetylcholine is effectively increased in the easily administered orally to children with JMG, but long-
neuromuscular junction, and acetylcholine is able to bind to term use can be harmful due to systemic side effects.1
the AChR for a longer period, leading to improved neuro- Children treated with prednisone demonstrated a good
muscular transmission. This test is performed with cardiac overall response in a retrospective study.47 Additionally,
monitoring and cardiopulmonary resuscitation equipment treatment with pre- dnisone has been found to have a
(including atropine) readily available owing to the risk of protective effect on the conversion from ocular to
bradycardia and asystole. During the test, attention should be generalized myasthenia.48,49 Treatment with prednisone
directed to a sign of myasthenia the patient exhibits (muscle appeared to provide control of OMG in a randomized
weakness, ptosis, etc) that can be readily monitored. Compar- clinical trial compared with placebo.50
ison to photographs can be useful. A small dose is given Azathioprine can be considered for use in myasthenia
initially to test for supersensitivity to edrophonium, followed gravis. Children who do not have an adequate response to
by escalating doses. Parasympathetic side effects including steroids and anticholinesterase treatment can be placed on
excessive salivation, hypotension, diarrhea, flushing, nausea, azathio- prine, and it can be used as a steroid-sparing
and sweating can occur. False negatives and false positives can agent.2,51-53 It should be kept in mind that azathioprine has a
occur with edrophonium testing as other diseases can respond delayed treatment effect from time of initiation of therapy,
to this test, or the patient may fail to respond to it.1 Neo- is potentially teratogenic, and is considered
stigmine is an alternative drug to edrophonium that can be carcinogenic.52,54
used in young children. Another alternative is to simply treat Other immunomodulatory medications can be
the patient with a trial of pyridostigmine and observe for considered for use in myasthenia gravis. Rituximab has
improvements in symptoms over time. been found to be effective in its treatment, improve
symptoms, and reduce the amount of steroids required for
treatment.55-57 Rituximab has also been found to be effective
Electrophysiological Testing for long-term treatment of refractory myasthenia gravis.58
Mycophenolate mofetil has also been found to be effective
Two methods of electrophysiological testing are primarily in the treatment of myasthenia gravis, but randomized
used for diagnosing myasthenia gravis. In one test, repetitive
nerve stimulation, trains of stimuli are delivered to a target control trials have not demonstrated the ability to taper
muscle, and quantitative analysis of the motor responses is steroids while on this agent.59-61 However, discontinuation
performed. A decremental response of 10% from the first to of mycophenolate mofetil in patients taking this medication
the fourth response is considered a positive result.43 This test does lead to more frequent myasthenia gravis
is more sensitive in patients who have generalized JMG in exacerbations.62 Tacrolimus has been used for the
comparison to those who have ocular JMG.44 Currently the treatment of myasthenia gravis as well, including JMG.63,64
“gold-standard” for testing for neuromuscular junction
disorders is single-fiber electromyography (sfEMG) performed
on the orbicularis oculi.43 The sfEMG is particularly useful in Other Medical Treatment Modalities
cases of suspected myasthenia gravis with negative serologies Intravenous immunoglobulin (IVIG) can be administered
as it has very high sensitivity for MG.45 Unfortunately, for myasthenia gravis in an effort to reduce the
traditional single-fiber EMG studies are difficult to perform on circulating autoantibodies by decreasing B-cell antibody
children due to discomfort and the length of the test. As an production and T-cell function. In a randomized
alternative, a less invasive test termed stimulated single-fiber controlled trial against placebo, IVIG was found to be
EMG has been developed that can be used in children.43,46 effective in improving myasthenia gravis symptoms.65
Therapy with IVIG has been used for patients with JMG,
but appeared to have better efficacy for short-term
Treatment treatments and exacerbations than for long-term
treatment.66,67 Plasmapheresis can also be used in
Acetylcholinesterase Inhibitors patients with myasthenia gravis to directly filter
Pyridostigmine is the main pharmacologic agent used in the autoantibodies and cytokines from the affected patient’s
treatment of myasthenia gravis, whether in children or serum. A comparison in patients with JMG between IVIG
adults. Edrophonium has too short a half-life to be of use in and plasmapheresis for maintenance therapy
long-term treatment of myasthenia gravis. Even with the demonstrated high response rates for both modalities,
appropriate usage and dosing of pyridostigmine, symptoms but a more consistent response with plasmapheresis. 68
and signs of myasthenia gravis may continue.1 However, pre- vious studies performed in adults have
Pyridostigmine may be combined with other agents such shown no signifi- cant difference between the 2
as immunosuppressives. Side effects of pyridostigmine are treatments.69 Consensus statements state that IVIG and
similar to those of edropho- nium described earlier. plasmapheresis are appro- priate for acute
exacerbations or crises only, and are not appropriate for
maintenance therapy unless patients are unable to
tolerate immunosuppressive agents or have refractory
myasthenia gravis.70
Thymectomy
References
Approximately 10%-15% of all adults with myasthenia 1. Andrews PI: Autoimmune myasthenia gravis in childhood. Semin
gravis are found to have a thymoma, and 30% of patients Neurol 24:101-110, 2004
who have a thymoma develop myasthenia gravis. 71,72 2. Mullaney P, Vajsar J, Smith R, Buncic JR: The natural history and
Children who have myasthenia gravis have rarely been ophthalmic involvement in childhood myasthenia gravis at the
found to harbor thymo- mas.22,73 However, thymectomy for Hospital for Sick Children. Ophthalmology 107:504-510, 2000
patients with seropositive JMG has been evaluated and has 3. Kinali M, Beeson D, Pitt MC, et al: Congenital myasthenic
been found to be associated with improved remission syndromes in childhood: Diagnostic and management challenges. J
rates.22,74 Therefore, thymectomy may have a role for Neuroimmunol 201-202:6-12, 2008
children who are unresponsive to medical therapy or who 4. Engel AG, Shen X, Selcen D, Sine SM: Congenital myasthenic
cannot undergo standard immunsuppression, although the syndromes: Pathogenesis, diagnosis, and treatment. Lancet Neurol
long-term effects of thymectomy in children are unclear.70 14:420-434, 2015
Thymectomy may protect patients with the ocular form of 5. Téllez-Zenteno JF, Hernández-Ronquillo L, Salinas V, et al:
JMG from undergoing generalization, and may improve Myasthenia gravis and pregnancy: Clinical implications and
ocular symptoms as well.9,24 A randomized trial of neonatal outcome. BMC Musculoskelet Disord 5:42, 2004
thymectomy for adult patients on prednisone for 6. Namba T, Brown SB, Grob D: Neonatal myasthenia gravis: Report
myasthenia gravis demonstrated a reduction in prednisone of two cases and review of the literature. Pediatrics 45:488-504,
dose require- ment and improvement in symptoms for 1970
those who under- went thymectomy.75 7. Evoli A: Acquired myasthenia gravis in childhood. Curr Opin Neurol
23:536-540, 2010
8. Liew WK, Kang PB: Update on juvenile myasthenia gravis. Curr
Opin Pediatr 25:694-700, 2013
Ophthalmic Outcomes 9. Gadient P, Bolton J, Puri V: Juvenile myasthenia gravis: Three case
reports and a literature review. J Child Neurol 24:584-590, 2009
Patients with JMG should be monitored for the 10. Pal J, Rozsa C, Komoly S, Illes Z: Clinical and biological heterogeneity
development of amblyopia and strabismus.76 Amblyopia of autoimmune myasthenia gravis. J Neuroimmunol 231:43-54, 2011
should be treated to prevent permanent vision loss. Up to 11. Tsivgoulis G, Dervenoulas G, Kokotis P, et al: Double seronegative
96% of patients with the ocular form of JMG will have myasthenia gravis with low density lipoprotein-4 (LRP4) antibodies
ptosis, and up to 88% will present with strabismus.28 presenting with isolated ocular symptoms. J Neurol Sci 346:328-330,
Amblyopia has been found in 21%- 50% of children with 2014
ocular JMG, but following properly diagnosis and treatment, 12. Zisimopoulou P, Evangelakou P, Tzartos J, et al: A comprehensive
that proportion of patients with amblyopia has been found analysis of the epidemiology and clinical characteristics of anti-LRP4 in
to decrease as low as 3%.24,25,28 Strabismus is more difficult myasthenia gravis. J Autoimmun 52:139-145, 2014
to treat in patients with ocular JMG, with its presence at 13. Tsao C, Mendell JR, Lo WD, et al: Myasthenia gravis and associated
final follow-up ranging from 15%- 57%.24,25 Ptosis may autoimmune diseases in childhood. J Child Neurol 15:767-769, 2000
remain in patients who have stable disease in 46%-66% of 14. Jarius S, Paul F, Franciotta D, et al: Neuromyelitis optica spectrum
patients.24,25 Upon stabilization of the underlying disease, disorders in patients with myasthenia gravis: Ten new aquaporin-4
surgical correction of the strabismus and ptosis can be antibody positive cases and a review of the literature. Mult Scler
considered, with a 56% long-term success rate found in 18:1135-1143, 2012
one retrospective study.77 Counseling should be given 15. Motobayashi M, Inaba Y, Nishimura T, et al: An increase in circulating
about realistic expectations of strabismus surgery and B cell-activating factor in childhood-onset ocular myasthenia gravis.
about the possibility of requiring multiple surgical Pediatr Neurol 52:404-409, 2015
interventions. 16. Motobayashi M, Nishimura T, Inaba Y: Usefulness of circulating B-
cell- activating factor in serial monitoring in childhood-onset ocular
myasthe- nia gravis. Pediatr Neurol 63:e1-e2, 2016
17. McGrogan A, Sneddon S, de Vries CS: The incidence of
Conclusions myasthenia gravis: A systematic literature review.
Neuroepidemiology 34:171-183, 2010
Pediatric myasthenia gravis can present as neonatal 18. Carr AS, Cardwell CR, McCarron PO, McConville J: A systematic review
myasthenia gravis (CMS, TNM) or during adolescence of population based epidemiological studies in myasthenia gravis.
(JMG). The latter is an autoimmune disease that can have a BMC Neurol 10:46, 2010
variable presentation, ranging from mild ophthalmic 19. Phillips LH, Torner JC, Anderson MS, Cox GM: The epidemiology of
symptoms, such as isolated fatigable ptosis, to myasthenic myasthenia gravis in central and western Virginia. Neurology 42:
crises involving the respiratory muscles, requiring 1888-1893, 1992
ventilator support. Making a formal diag- nosis of the 20. Zhang X, Yang M, Xu J, et al: Clinical and serological study of
disease can be difficult, but when suspicion is raised for a myasthenia gravis in HuBei Province, China. J Neurol Neurosurg
fatigable deficit, appropriate diagnostic testing can be Psychiatry 78: 386-390, 2007
pursued. Treatments should be tailored to the subtype of 21. Chiu H-C, Vincent A, Newsom-Davis J, et al: Myasthenia gravis:
disease and the severity of illness. Variable responses to Population differences in disease expression and acetylcholine
receptor antibody titers between Chinese and Caucasians. Neurology
medications and surgical interventions can be seen
37:1854-1857, 1987
between patients. Children with involvement of the
22. Castro D, Derisavifard S, Anderson M, et al: Juvenile myasthenia
extraocular mus- culature should be followed by a pediatric gravis: A twenty-year experience. J Clin Neuromusc Dis 14:95-102,
ophthalmologist to monitor for ophthalmic complications 2013
associated with JMG. 23. Nemet AY, Segal O, Mimouni M, Vinker S: Associated morbidity of
pediatric ptosis—a large, community based case-control study.
Graefes Arch Clin Exp Ophthalmol 252:1509-1514, 2014
24. Pineles SL, Avery RA, Moss HE, et al: Visual and systemic outcomes in
pediatric ocular myasthenia gravis. Am J Ophthalmol 150:453-459,
2010
25. Ortiz S, Borchert M: Long-term outcomes of pediatric ocular
myasthenia gravis. Ophthalmology 115:1245-1248, 2008
26. Nagia L, Lemos J, Abusamra K, et al: Prognosis of ocular myasthenia
gravis: Retrospective multicenter analysis. Ophthalmology 122:1517
1521, 2015
27. Bever CT, Aquino AV, Penn AS, et al: Prognosis of ocular myasthenia. Ann Neurol 14:516-519, 1983
28. Kim JH, Hwang JM, Hwang YS, et al: Childhood ocular myasthenia gravis. Ophthalmology 110:1458-1462, 2003
29. McCreery KM, Hussein MA, Lee AG, et al: Major review: the clinical spectrum of pediatric myasthenia gravis: blepharoptosis, ophthalmoplegia and
strabismus. A report of 14 cases. Binocul Vis Strabismus Q 17: 181-186, 2002
30. Jaretzki A 3rd, Barohn RJ, Enrstoff RM, et al: Myasthenia gravis: Recommendations for clinical research standards. Task force of the Medical
Scientific Advisory Board of the Myasthenia Gravis Foundation of America. Neurology 55:16-23, 2000
31. Golnik KC, Pena R, Lee AG, Eggenberger ER: An ice test for the diagnosis of myasthenia gravis. Ophthalmology 106:1282-1286, 1999
32. Sethi KD, Rivner MH, Swift TR: Ice pack test for myasthenia gravis. Neurology 37:1383-1385, 1987
33. Odel JG, Winterkorn JM, Behrens MM: The sleep test for myasthenia gravis. A safe alternative to Tensilon. J Clin Neuroophthalmol 11: 288-292,
1991
34. Kubis KC, Danesh-Meyer HV, Savino PJ, Sergott RC: The ice test versus the rest test in myasthenia gravis. Ophthalmology 107:1995-1998, 2000
35. Della Marina A, Trippe H, Lutz S, Schara U: Juvenile myasthenia gravis: Recommendations for diagnostic approaches and treatment. Neuropedi-
atrics 45:75-83, 2014
36. Anlar B, S¸enbil N, Köse G, Değerliyurt A: Serological follow-up in juvenile myasthenia: Clinical and acetylcholine receptor antibody status of patients
followed for at least 2 years. Neuromuscul Disord 15:355-357, 2005
37. VanderPluym J, Vajsar J, Jacob FD, et al: Clinical characteristics of pediatric myasthenia: A surveillance study. Pediatrics 132:e939-e944, 2013
38. Zhou L, McConville J, Chaudhry V, et al: Clinical comparison of muscle- specific tyrosine kinase (MuSK) antibody-positive and -negative myas-
thenic patients. Muscle Nerve 30:55-60, 2004
39. Skjei KL, Lennon VA, Kuntz NL: Muscle specific kinase autoimmune myasthenia gravis in children: A case series. Neuromuscul Disord 23: 874-882, 2013
40. Evoli A, Tonali PA, Padua L, et al: Clinical correlates with anti-MuSK antibodies in generalized seronegative myasthenia gravis. Brain 126: 2304-2311, 2003
41. Pevzner A, Schoser B, Peters K, et al: Anti-LRP4 autoantibodies in AChR- and MuSK-antibody-negative myasthenia gravis. J Neurol 259:427-435, 2012
42. Binks S, Vincent A, Palace J: Myasthenia gravis: A clinical-immunological update. J Neurol 263:826-834, 2016
43. Pitt M: Neurophysiological strategies for the diagnosis of disorders of the neuromuscular junction in children. Dev Med Child Neurol 50:328-333,
2008
44. Afifi AK, Bell WE: Tests for juvenile myasthenia gravis: Comparative diagnostic yield and prediction of outcome. J Child Neurol 8:403-411, 1993
45. Padua L, Stalberg E, LoMonaco M, et al: SFEMG in ocular myasthenia gravis diagnosis. Clin Neurophysiol 111:1203-1207, 2000
46. Verma S, Lin J: Stimulated jitter analysis for the evaluation of neuro- muscular junction disorders in children. Muscle Nerve 53:471-472, 2016
47. Batocchi AP, Evoli A, Palmisani MT, et al: Early-onset myasthenia gravis: Clinical characteristics and response to therapy. Eur J Pediatr 150:66-68, 1990
48. Kupersmith MJ, Latkany R: Development of generalized disease at two years in patients with ocular myasthenia gravis. Arch Neurol 60:243-248,
2003
49. Kupersmith MJ: Ocular myasthenia gravis: Treatment successes and failures in patients with long-term follow-up. J Neurol 256:1314-1320, 2009
50. Benatar M, McDermott MP, Sanders DB, et al: Efficacy of prednisone for the treatment of ocular myasthenia (EPITOME): A randomized, con-
trolled trial. Muscle Nerve 53:363-369, 2016
51. Ashraf VV, Taly AB, Veerendrakumar M, Rao S: Myasthenia gravis in children: A longitudinal study. Acta Neurol Scand 114:119-123, 2006
52. Ionita CM, Acsadi G: Management of juvenile myasthenia gravis. Pediatr Neurol 48:95-104, 2013
53.
Palace J, Newsom-Davis J, Lecky B: A randomized double-blind trial of prednisone alone or with azathioprine in myasthenia gravis: Myasthenia Gravis Study
Group. Neurology 50:1778-1783, 1998

54. Haines SR, Thurtell MJ: Treatment of ocular myasthenia gravis. Curr Treat
Options Neurol 14:103-112, 2012

55. Wylam ME, Anderson PM, Kuntz NL, Rodriguez V: Successful treatment of refractory myasthenia gravis using rituximab: A pediatric case report. J
Pediatr 143:674-677, 2003
56. Stieglbauer K, Topakian R, Schäffer V, Aichner FT: Rituximab for
myasthenia gravis: Three case reports and review of the literature. J Neurol Sci 280:120-122, 2009

57. Koul R, Al Futaisi A, Abdwani R: Rituximab in severe seronegative juvenile


myasthenia gravis: Review of the literature. Pediatr Neurol 47:209-212, 2012

58. Robeson KR, Kumar A, Keung B, et al: Durability of the rituximab


response in acetylcholine receptor autoantibody-positive myasthenia gravis. JAMA Neurol 74:60-66, 2017

59. Hehir MK, Burns TM, Alpers J, et al: Mycophenolate mofetil in AChR-
antibody-positive myasthenia gravis: Outcomes in 102 patients. Muscle Nerve 41:593-598, 2010

60. Muscle Study Group. A trial of mycophenolate mofetil with prednisone as


initial immunotherapy in myasthenia gravis. Neurology 71:394-399, 2008

61. Sanders DB, Hart IK, Mantegazza R, et al: An international, phase III, randomized trial of mycophenolate mofetil in myasthenia gravis. Neurol-
ogy 71:400-406, 2008
62. Oskarsson B, Rocke DM, Dengel K, Richman DP: Myasthenia
gravis exacerbation after discontinuing mycophenolate. Neurology 86: 1159-1163, 2016

63. Minami N, Fujiki N, Doi K, et al: Five-year follow-up with low-dose


tacrolimus in patients with myasthenia gravis. J Neurol Sci 300:59-62, 2011

64. Mori T, Mori K, Suzue M, et al: Effective treatment of a 13-year-old boy with steroid-dependent ocular myasthenia gravis using tacrolimus.
Brain Dev 35:445-448, 2013
65. Zinman L, Ng E, Bril V: IV immunoglobulin in patients with myasthenia
gravis. Neurology 68:837-841, 2007

66. Selcen D, Dabrowski ER, Michon AM, Nigro MA: High-dose intravenous immunoglobulin therapy in juvenile myasthenia gravis. Pediatr Neurol
22:40-43, 2000
67. Feasby T, Banwell B, Benstead T, et al: Guidelines on the use of
intravenous immune globulin for neurologic conditions. Transfus Med Rev 21:S57-S107, 2007

68. Liew WK, Powell CA, Sloan SR, et al: Comparison of plasmapheresis and
intravenous immunoglobulin as maintenance therapies for juvenile myasthenia gravis. JAMA Neurol 71:575-580, 2014

69. Gajdos P, Chevret S, Toyka KV: Intravenous immunoglobulin for


myasthenia gravis. Cochrane Database Syst Rev 12:CD002277, 2012

70. Sanders DB, Wolfe GI, Evoli A, et al: International consensus guidance for management of myasthenia gravis. Neurology 87:419-425, 2016
71. Marx A, Pfister F, Schalke B, et al: The different roles of the thymus in the pathogenesis of the various myasthenia gravis subtypes. Autoimmun
Rev 12:875-884, 2013
72. Gilhus NE, Verschuuren JJ: Myasthenia gravis: Subgroup classification and therapeutic strategies. Lancet Neurol 14:1023-1036, 2015
73. Nikolic DM, Nikolic AV, Lavrnic DV, et al: Childhood-onset myasthenia gravis with thymoma. Pediatr Neurol 46:329-331, 2012
74. Heng HS, Lim M, Absoud M, et al: Outcome of children with
acetylcholine receptor (AChR) antibody positive juvenile myasthenia gravis following thymectomy. Neuromuscul Disord 24:25-30, 2014

75. Wolfe GI, Kaminski HJ, Aban IB, et al: Randomized trial of thymectomy in
myasthenia gravis. N Engl J Med 375:511-522, 2016

76. Emoto Y, Emoto H, Fujie W, Wakakura M: Prolonged constant eye misalignment leads to failure to develop binocular vision in childhood ocular
myasthenia gravis. J Pediatr Ophthalmol Strabismus 46:358-361, 2009
77. Peragallo JH, Velez FG, Demer JL, Pineles SL: Long-term follow-up of
strabismus surgery for patients with ocular myasthenia gravis. J Neuro- ophthalmol 33:40-44, 2013
CMScri
Member of a medical scheme? Know your guaranteed benefits!

pt Issue 5 of 2017

Myasthenia
Gravis
Myasthenia Gravis (MG) is a Chronic Autoimmune Neuromuscular condition that causes weakness in cer- tain
muscles of the body. MG is caused by an error in the transmission of nerve impulses to the muscles. In MG
the immune system makes antibodies that mistakenly attack the connections between nerves and muscles
preventing muscles from contracting and resulting in weakness.

What does
autoimmune
mean? The immune
system (the body’s in-
fection-fighting technique)
normally makes proteins
called “antibodies,” which
help to prevent or fight
infec- tion.

Antibodies are proteins


made by the body’s immune
system when it de- tects
harmful substances.
Antibodies may be produced
when the immune system
mistakenly considers
healthy tissue as harmful
substance.

What is the neuromuscular


junction?
The Neuromuscular junction is the
meeting point of a nerve cell and
the muscle it controls.
Communication
happens between the nerve and muscle fibers through
the nerve cells called neurons. Due to this communica- and resulting in weakness. The diagram on the next page
tion or transmission of signal, the muscle is able to con- shows the antibodies attacking the junction between nerves
tract or relax. When the muscles contract, Acetylcholine and muscle.
gets released by the nerve cells. Acetylcholine acts as a
chemical messenger that causes our skeletal muscles to Some people may have Myasthenia Gravis that is not caused
contract. The diagram above right shows the neuromus- by antibodies blocking Acetylcholine or the mus- cle-
cular junction. specific receptor tyrosine kinase. This type of condition is
called antibody-negative Myasthenia Gravis.
What causes Myasthenia Gravis?
MG is caused by an error in the transmission of nerve im- The role of thymus gland in myasthenia gravis
pulses to the muscles. In MG the immune system makes The thymus gland, lies in the chest area beneath the
antibodies that mistakenly attack the connections between breastbone (sternum), and plays an important role in the
nerves and muscles preventing muscles from contracting development of the immune system in early life. Its cells
form part of the body’s normal immune system. The gland

Figure 1: The neuromuscular


junction
• Jaw muscles
• Arm or leg muscles
• Muscles that help with breathing

What are the symptoms of Myasthenia


Gravis? The main symptom is muscle weakness. It
can come and go, and is often worse later in the
day. This can result in:
• Ptosis (Droopy eyelids).
• Diplopia (Blurry vision or double vision).
• Trouble chewing food - The jaw muscles might
feel tired about halfway through a meal.
• Trouble swallowing.
• Dysarthria (Trouble talking) – A person might
speak in a lower voice than usual, or sound like
he or she has a cold or stuffy nose.
• Loss of expression on the face.
• Head that feels heavy or drops forward.
• Trouble breathing – A person might feel short
of breath, take extra breaths, or feel like it
takes a lot of effort to breathe.
• Weakness – It might be hard to lift the arms or
Figure 2: Antibodies attacking the junction between nerves and
muscle.
legs, open the fingers, or lift a foot.

Who gets Myasthenia Gravis?


Myasthenia Gravis occurs in all ethnic groups and both genders.
It most commonly affects young adult women (under 40) and
older men (over 60), but can also occur at any age. The
condition is not directly inherited nor is it contagious.
is somewhat larger in infants, grows gradually until puberty, and
then gets smaller and is replaced by fat with age. In adults with
Myasthenia Gravis, the thymus gland remains large and is What is covered under PMB level of care?
abnormal. Some individuals with Myasthenia Gravis develop PMBs refer to the benefits as stated in Section 29 (1) (o) of the
thymomas (tumors of the thymus gland). Medical Schemes Act, No. 131 of 1998 (the Act) Myasthenia
Gravis is a PMB condition under Diagnosis and Treatment Pair
The relationship between the thymus gland and Myasthenia (DTP) code 513A.
Gravis is not yet fully understood. Scientists believe that the
thymus gland may give incorrect instructions to developing This DTP refers to Myasthenia gravis as muscular dystro- phy;
immune cells, ultimately resulting in autoimmunity. neuro-myopathies NOS. The treatment component specified for
this DTP according to the PMB Regulations is Initial diagnosis;
initiation of medical management; therapy for acute
What happens if I have Myasthenia Gravis? complications and exacerbations. The following should
The hallmark of Myasthenia Gravis is muscle weakness that therefore be paid according to the PMB Regulations:
increases during periods of activity and improves after periods of
rest. Certain muscles such as those that con- trol eye and eyelid
Diagnosis
movement, facial expression, chewing, talking, and swallowing are
The doctor will record the patient’s medical history, do a
often, but not always, involved in the disorder. The muscles that
physical and detailed neurological examination to under- stand
control breathing and neck and limb movements may also be
the symptoms. If the doctor suspects Myasthenia Gravis, several
affected.
tests can be performed to confirm the di- agnosis. Not all
patients qualify for all the tests mentioned below. The
These include:
investigations are done subject to the discre- tion of the treating
• Muscles in the eyelids and around the eyes – If doctor.
MG only affects these muscles, doctors call it
“ocular my- asthenia gravis.” About half of all
people with MG have this type.
2 CMScript
5/2017
The tests can include: hyperplasia of the thymus gland.
• Blood tests for certain antibodies that are found • Treatment options also include
in peo- ple with MG such as acetylcholine methotrexate, myco- phenolate mofetil and
receptor antibodies. cyclosporine.
• Electrical tests of nerves and muscles –
Repetitive nerve stimulation.This is a nerve As the PMB Regulations stipulate initiation of treatment, the
chronic use of the abovementioned treatments is not included
conduction study in which doctors attach and the medical scheme does not have to fund the chronic
electrodes to your skin over the muscles to be medicine in full.
tested. Doctors send small pulses of electricity
through the electrodes to measure the nerve’s
ability to send a signal to your muscle. To di-
agnose Myasthenia Gravis, doctors will test the
nerve many times to see if its ability to send
signals worsens with fatigue.
• Imaging tests, such as Computerized
Tomography (CT) or Magnetic Resonance
Imaging (MRI) scans – Imaging tests can show
changes, including a tumor on the thymus gland.
• Ice pack test – During this test, a doctor puts a
cool pack on the eyelids. If the eyelids open
better after rest- ing under the cool pack, this
could be a sign of MG.
• Pulmonary function testing - which measures
breathing strength, helps to predict whether
respiration may fail and lead to a Myasthenia
crisis.

How is Myasthenia Gravis treated?


Initiation of treatments, which may include:
• Medicines to treat muscle weakness, such as
pyr- idostigmine.
• Medicines that treat the immune system over
time, such as prednisone or azathioprine. These
medications im- prove muscle strength by
suppressing the production of abnormal
antibodies.
• Fast-acting immune system treatments, such as:
• A medicine called “intravenous immune
globulin” (IVIG) – high-dose intravenous
immune globulin, which temporarily
modifies the immune system by infusing
antibodies from donated blood.
• Plasma exchange (also called
“plasmapheresis”)
–a procedure in which serum containing the ab- normal
antibodies is removed from the blood while cells are
replaced.
• Surgery to remove the thymus gland in patients
with thymoma (tumor of the thymus gland /
abnormal tis- sue of the thymus gland) or

CMScript 3
Therapy for exacerbations and acute complications such as pneumonia
and respiratory failure must however be funded as PMB level of care
by the medical scheme. What else should I know about Myasthenia
Gravis? People who have MG that affects more than just the
These therapies may be used to help individuals during especially eyes can have serious problems if they get the flu or pneumonia.
difficult periods of weakness. A Neurologist will determine which For this reason, it is especially important that they get the flu
treatment option is best for each individ- ual depending on the severity vaccination every year and the pneumonia vaccine as per the
of the weakness, the kind of muscles are affected, as well as the national vaccination guideline.
individual’s age and other associated medical problems.
Some medicines can make MG worse. Talk to your doctor or nurse
If MG attacks the muscles that help with breathing, it can cause severe
before you take any medicines, including over-the- counter
breathing problems. These patients are usu- ally treated in the
medicines. If you get a prescription for a new med- icine, ask if it is
Intensive Care Unit (also called the “ICU”) and may require assisted
safe to take when you have MG.
ventilation.

Children with MG can take some of the same medicines prescribed for References
adults, at age appropriate doses. However, some medicines used to
1. Choi, D. E., Hobson-Webb, L.D., & Juel, V.C.l.
treat MG can cause more serious problems in children if used for a long
time. Surgery to re- move the thymus gland is safe for children and (2014). Do acetylcholine receptor and striated
often works well. muscle antibod- ies predict the presence of
thymoma in patients with myasthenia gravis.
What is a Myasthenic crisis? Muscle Nerve, 49:30.
A Myasthenic Crisis occurs when the muscles that control breathing
weaken to the point that ventilation is inadequate, creating a medical 2. Golnik, K.C., Pena, R., Lee, A.G. & Eggenberger,
emergency and requiring a respirator for assisted breathing. E.R. (1999). An ice test for the diagnosis of
Respiratory failure is included in the PMBs and the medical scheme
must fund the medical treat- ment, oxygen and ventilation in full. myasthenia gra- vis. Ophthalmology, 106:1282.
3. Krendel, D.A., Sanders, D.B., Massey, J.M. (1987).

Single fiber electromyography in chronic progressive external


ophthalmoplegia. Muscle Nerve, 10:299.
4. Mahadeva, B., Phillips, L.H. 2nd & Juel, V.C.
(2008). Autoimmune disorders of
neuromuscular transmission. Semin Neurol,
28:212.
5. Matell, G. (1987). Immunosuppressive drugs:
azathio- prine in the treatment of myasthenia
gravis. Ann N Y Acad Sci, 505:589.
6. Meriggioli, M.N., Sanders, D.B. (2009).
Autoimmune myasthenia gravis: emerging
clinical and biological heterogeneity. Lancet
Neurol, 8:475.
7. Milone, M., Monaco, M.L., Evoli, A. (1993).
Ocular my- asthenia: diagnostic value of single
fibre EMG in the orbicularis oculi muscle. Journal
of Neurol Neurosurg Psychiatry, 56:720.
WHAT ARE PRESCRIBED MINIMUM BENEFITS?
8. Muscle Study Group. (2008). A trial of mycophenolate
mofetil with prednisone as initial immunotherapy in my- Prescribed Minimum Benefits (PMBs) are defined by
asthenia gravis. Neurology, 71:394. law. They are the minimum level of
9. National Institute of neurological disorders and diagnosis,treatment, and care that your medical
stroke: Myasthenia Gravis scheme must cover – and it must pay for your PMB
10. Pascuzzi, R.M. (2003). The edrophonium test. condition/s from its risk pool and in full. There are
Semin Neurol, 23:83. medical interventions available over and above those
11. Sanders, D.B., Hart, I.K. & Mantegazza, R. (2008). prescribed for PMB conditions but your scheme may
An international, phase III, randomized trial of choose not to pay for them. A desig- nated service
mycophe- nolate mofetil in myasthenia gravis. provider (DSP) is a healthcare provider (e.g. doctor,
pharmacist, hospital) that is your medical scheme’s
first choice when you need treatment or care for a
4 CMScript PMB condition. You can use a non-DSP voluntarily or
use a non-DSP, you may have to pay a portion of the
bill as a co-payment. PMBs include 270 serious health
conditions, any emergency condition, and 25 chronic
diseases; they can be found on our website
Neurology, 71:400.
12. Sanders, D.B., Wolfe, G.I. & Benatar, M. (2016).
Inter- national consensus guidance for
management of myas- thenia gravis: Executive
summary. Neurology, 87:419.
13. Sethi, K.D., Rivner, M.H. & Swift, T.R. (1987). Ice
pack test for myasthenia gravis. Neurology,
37:1383.
14. Vernino, S. & Lennon, V.A. (2004). Autoantibody
pro- files and neurological correlations of
thymoma. Clin Cancer Res, 10:7270.
15. Figure 1 -
http://www.mayoclinic.org/-/media/kcms/
gbs/patient-
consumer/images/2013/08/26/10/28/
ds00375_im00332_r7_receptorsthu_jpg.jpg
[Ac- cessed 12 June 2017]
16. Figure 2 -
http://www.chop.edu/sites/default/files/my-
asthenia-gravis-neuromuscular-junction-
illustration- 773x949.png [Accessed 12 June
2017]
T he NE W E NGL A ND JOU R NA L o f M E DIC INE

REVIEW ARTICLE

Dan L. Longo, M.D., Editor

Myasthenia Gravis

From the Department of Clinical Medi-


cine, University of Bergen, and the Depart-
M Nils E. Gilhus, M.D.

YASTHENIA GRAVIS IS AN AUTOIMMUNE DISEASE IN WHICH ANTI- bodies bind to


acetylcholine receptors or to functionally related molecules in the postsynaptic membrane at
ment of Neurology, Haukeland University
the neuromuscular junction. The anti-
Hospital — both in Bergen, Norway. Ad-
dress reprint requests to Dr. Gilhus at the bodies induce weakness of skeletal muscles, which is the sole disease manifesta- tion.1-3 The weakness
Department of Neurology, Haukeland Uni- can be generalized or localized, is more proximal than dis- tal, and nearly always includes eye
versity Hospital, 5021 Bergen, Norway, or at muscles, with diplopia and ptosis.2 The pattern of involvement is usually symmetric, apart from the
nils.gilhus@uib.no. eye involvement, which is often markedly asymmetric and involves several eye muscles. The
N Engl J Med 2016;375:2570-81.
weakness typi- cally increases with exercise and repetitive muscle use (fatigue) and varies over the
DOI: 10.1056/NEJMra1602678 course of a day and from day to day, often with nearly normal muscle strength in the morning.
Copyright © 2016 Massachusetts Medical Society. With an annual incidence of 8 to 10 cases per 1 million persons and a preva- lence of 150 to
250 cases per 1 million,4 myasthenia gravis and its various sub- groups are the major diseases that
affect the neuromuscular junction. The Lambert– Eaton myasthenic syndrome and neuromyotonia are
additional, rare, presynaptic autoantibody disorders characterized by skeletal-muscle dysfunction.5
Congenital myasthenic syndromes and toxin-induced conditions (e.g., botulism) can also affect the
neuromuscular junction and lead to muscle weakness. This review focuses on new diagnostic tests for
myasthenia gravis, updated treatment algorithms, and individualization of therapy according to
biomarkers.
The diagnosis of myasthenia gravis is confirmed by the combination of relevant symptoms and
signs and a positive test for specific autoantibodies.6 Antibodies against acetylcholine receptors,
muscle-specific kinase, and lipoprotein receptor– related protein 4 (LRP4) are specific and sensitive
for the detection of myasthenia gravis, define disease subgroups, and point to pathogenic variations
among these subgroups. The localization of the antigens at the neuromuscular junction and in skeletal
muscle is shown in Figure 1. The disease-inducing potential of the anti- bodies depends on the
epitope, binding pattern, IgG subclass, antibody cross-linking capacity, antibody concentration, and
access of antibody to the muscle end plate.7 In antibody-negative cases, neurophysiological tests and a
characteristic re- sponse to therapy secure the diagnosis.8 An ice-pack test that reverses ptosis sup-
ports the diagnosis. Thymic status should be determined by means of mediastinal imaging.9 The main
value of such imaging is to detect a thymoma; this imaging is neither sensitive nor specific for the
identification of thymic hyperplasia. Sup- plementary antibody tests can provide further help in
characterizing the thymus.10 Symptomatic, immunoactive, and supportive approaches to therapy have a
very good effect, and the prognosis regarding muscle strength, functional abilities, quality of life,
and survival is generally good.11,12 Therapy should be aimed at full or nearly full pharmacologic
remission (i.e., the absence of myasthenic symptoms
and signs while the patient is receiving therapy).
M YASTHENIA G R AVIS

MITOCHONDRION

Synaptic vesicle

MOTOR NEURON

Acetylcholinesterase
SYNAPTIC CLEFT
Inhibition improves
symptoms of
myasthenia gravis Acetylcholin
Agrin e
Antibody
targets

Antibody target
MuSK

Acetylcholine
receptor

LRP4 Dok7
Rapsyn

Antibodies as biomarkers

Ryanodine
receptor

MUSCLE CELL

SARCOPLASMIC RETICULUM

Antibodies as biomarkers

Titin

SARCOMERE

Figure 1. Neuromuscular Junction and Key Elements for the Pathogenesis of Myasthenia Gravis.

Neuromuscular transmission involves release of presynaptic acetylcholine, which binds to acetylcholine receptors in the
postsynaptic membrane. The receptors interact with several other proteins in the membrane, including Dok7 and rapsyn. Mutant
Dok7 and rapsyn are important in the development of congenital myasthenia. Antibodies against acetylcholine receptors, as well
as antibodies against muscle-specific kinase (MuSK) and lipoprotein receptor–related peptide 4 (LRP4), induce myasthenic weakness.
Antibodies against the intramuscular proteins titin and ryanodine receptor are relevant biomarkers in some subgroups of
myasthenia gravis. Acetylcholine is degraded by local acetylcholinesterase, and acetylcholinesterase inhibition leads to
symptomatic improvement in patients with myas- thenia gravis.

The New England Journal of Medicine


Downloaded from nejm.org at TULANE UNIV on January 16, 2019. For personal use only. No other uses without permission.
Copyright © 2016 Massachusetts Medical Society. All rights reserved.
N ENGL J MED 375;26 NEJM.ORG DECEMBER 29, 2016 2571

The New England Journal of Medicine


Downloaded from nejm.org at TULANE UNIV on January 16, 2019. For personal use only. No other uses without permission.
Copyright © 2016 Massachusetts Medical Society. All rights reserved.
T he NE W E NGL A ND JOU R NA L o f M E DICI NE

C L I N IC A L A ND P A T HO GE N IC the immune system and probably the risks of


VA R I A N T S autoimmune disease; late-onset disease is asso-
ciated with HLA-DR2, HLA-B7, and HLA-DRB1
17,18
Variants of myasthenia gravis are defined on the basis 15.01. Thymic status and HLA pattern repre- sent
of autoimmune and antibody disease mech- anisms, strong subgroup markers, probably point- ing directly
target molecules of skeletal muscle, thymic status, to variation in pathogenic pathways. Early-onset
genetic characteristics, response to therapy, and myasthenia gravis is three times as likely to be
disease phenotype. Patients with myasthenia gravis diagnosed in females as it is in males, whereas
should always be subgrouped on the basis of all these males slightly outnumber fe- males in the late-
variables (Fig. 2A) and should be assigned to only onset group. Coexisting auto-
one subgroup. To
combine disparate clinical and nonclinical fea-
tures in the classification of individual patients is A Myasthenia Gravis Subgroups
a challenge (Table 1). Subgroups influence thera- peutic decisions and prognosis.
In 15% of all patients with myasthenia gravis,
symptoms and signs are confined to ocular
muscles. Only half of patients with ocular myas- thenia
Ocular
gravis have detectable muscle antibod- ies.13 Ptosis Early onset
and diplopia are common initial symptoms, but the
disease remains restricted to ocular muscles in only a Seronegative
minority of patients. In 90% of patients who
continue to have purely ocular myasthenia gravis 2
years after the start of symptoms, the disease will MuSK
persist as a focal eye-muscle weakness and never LRP4
Late onset
become general- ized. Myasthenia gravis with
Thymoma
muscle-specific kinase antibodies is not manifested
as ocular myasthenia, whereas both acetylcholine
receptor and LRP4 antibodies can be found in the
ocular subgroup.2 The presence of muscle
antibodies increases the risk of subsequent B Coexisting Conditions
generalized disease.
Ten percent of patients with myasthenia gra-
vis have a thymoma, and the prevalence increases with
Thymoma
increasing age. Two thirds of patients with
myasthenia gravis have generalized early-onset or
late-onset disease and no thymoma. Among patients
who have myasthenia gravis with acetyl- choline Autoimmunity
Therapy- Myasthenia
induced gravis
receptor antibodies, the age at onset has a bimodal disorders
pattern, supporting the use of a cutoff age of 50 years
to distinguish between early- onset and late-onset
disease.14 Juvenile myasthe- nia gravis, which is Cardiac
included in the early-onset group and is defined as Unrelated
disease
an onset before the age of 15 years, is much more disorders
common in East Asian populations than in whites.15,16
Early-onset myas- thenia gravis tends to be
characterized by thymic hyperplasia, whereas thymic
atrophy is charac- teristic of late-onset disease. Early-
onset myasthe- nia gravis is associated with HLA-DR3,
HLA-B8, and non-HLA genes that are known to Figure 2. Subgroups of Myasthenia Gravis and Coexisting
influence Conditions.
Panel A shows myasthenia gravis subgroups defined on
the basis of clinical, antibody, and thymic features. MuSK
2572 denotes muscle-specific kinase, and LRP4 lipo- protein
N ENGL J MED 375;26 NEJM.ORG DECEMBER 29, 2016
receptor–related protein 4. As shown in Panel B, patients
with myasthenia gravis commonly have coexist- ing
conditions that are related to their disease (especially
thymoma and other autoimmune conditions), induced by
therapy, or unrelated to their disease.
M YASTHENIA G R AVIS

Table 1. Features of Myasthenia Gravis Subgroups.*

Subgroup Antibody Age at Onset Thymus


Early onset Acetylcholine receptor <50 yr Hyperplasia common
Late onset Acetylcholine receptor ≥50 yr Atrophy common
Thymoma Acetylcholine receptor Any age Lymphoepithelioma
Muscle-specific kinase Muscle-specific kinase Any age Normal
LRP4 LRP4 Any age Normal
Seronegative None detected Any age Variable
Ocular Variable Any age Variable

* LRP4 denotes lipoprotein receptor–related protein 4.

immune disorders are more common in early-


antibodies are associated with any proven thymic
onset disease than in late-onset disease.19 disease. A few patients with muscle-specific ki- nase
Titin antibodies, which occur primarily in pa- tients or LRP4 antibodies in combination with
with a thymoma and late-onset myasthe- nia gravis, in acetylcholine receptor antibodies have been de-
addition to acetylcholine receptor antibodies,10,20 scribed.1,6 Such patients should be classified ac-
have been shown to be a marker for severe disease. cording to the muscle-specific kinase or LRP4
Ryanodine receptor antibod- ies, which are present antibodies.
in 70% of patients with a thymoma and myasthenia In some patients with myasthenia gravis, no serum
gravis and in 14% of those with late-onset myasthenia antibodies against neuromuscular junction proteins can
gravis,10 are a marker for more severe disease but be detected. After standard testing with
have no disease-modifying effects. Kv1.4 antibodies commercially available kits, 10 to 15% of patients
are detected in 10 to 20% of patients with acetylcho- remain seronegative. Cell-based assays for antibody
line receptor antibodies.21,22 The disease pheno- type detection are more sensitive than serum tests
does not differ between early-onset and late-onset because the antigens expressed on cell membranes
myasthenia gravis. can be clustered and maintain their natural
Myasthenia gravis with muscle-specific kinase conformation.27 Such cell-based assays have been
antibodies accounts for 1 to 10% of cases.23 This developed for acetylcholine re- ceptor, muscle-
disorder is more common in the Mediterranean area specific kinase, and LRP4 anti- bodies.1,6 One third of
of Europe than in northern Europe and is also patients with generalized myasthenia gravis who are
more common in the northern regions of East Asia seronegative on stan- dard testing are seropositive on
than in the southern regions.24 The reason for this cell-based test- ing. The seronegative group probably
variation is thought to be a ge- netic predisposition includes some patients with acetylcholine receptor,
rather than environmental factors. Patients with muscle- specific kinase, or LRP4 antibodies that are
myasthenia gravis and muscle-specific kinase not detected because of insufficient test sensitivity.
antibodies, as compared with patients without these Some patients may have pathogenic antibodies
antibodies, have more severe weakness, sometimes against other postsynaptic membrane antigens. These
with muscle atro- phy, and have marked symptoms antigens interact with acetylcholine recep- tors. Some
from facial and bulbar muscles. Limb weakness and patients may have disease that is not mediated by
ocular weak- ness are less common and fluctuations in antibodies.
muscle strength are less pronounced than in Agrin antibodies, in the absence of other
disease characterized by acetylcholine receptor muscle antibodies, have been found in a minor- ity of
antibodies. LRP4 antibodies are present in 1 to 3% of patients with myasthenia gravis.28 These antibodies
all patients with myasthenia gravis.25,26 Such patients seem to be specific for myasthenia gravis. Agrin
tend to have only mild-to-moderate symptoms. has regulatory properties in the postsynaptic
Neither cases of myasthenia gravis with LRP4 membrane and is linked to neuro- muscular
antibodies nor those with muscle-specific kinase transmission, but so far, a pathogenic effect of agrin
antibodies has not been estab-

N ENGL J MED 375;26 NEJM.ORG DECEMBER 29, 2016 2573


T he NE W E NGL A ND JOU R NA L o f M E DICI NE

lished. Collagen Q and cortactin antibodies have been clinical heart disease and heart dysfunction are very
detected in some patients.1,29 The specificity of these rare. In population-based studies, myasthenia gravis
antibodies for myasthenia gravis has been has not been associated with an increase in
questioned. mortality related to heart disease.36 Functional
In seronegative patients with myasthenia gra- vis, imaging studies have shown minor and sub- clinical
the diagnosis should be reevaluated, and anti- body dysfunction.37 Myocarditis in myasthenia gravis is
tests should be repeated after 6 to 12 months. Before associated with Kv1.4 muscle antibod- ies.22 Antibodies
sensitive cell-based assays are included in clinical against acetylcholine receptor, muscle-specific kinase,
practice, standard procedures for these assays, as well and LRP4 do not cross- react with heart muscle, in
as their disease specificity, need to be defined. contrast to nonjunc- tional antibodies against Kv1.4,
titin, and ryano- dine receptor.38
C O e x I S T I NG D I S OR DER S For the most part, patients with myasthenia
gravis do not seem to have any clinically relevant
Coexisting conditions are common in patients with increase in the risk of cancer.39,40 The exception is
myasthenia gravis and should always be considered the subgroup of patients with thymoma. The
(Fig. 2B). Approximately 15% of pa- tients have a increased cancer risk among patients with thy- moma
second autoimmune disease,19,30 which occurs most is the same whether or not they have myasthenia
frequently in patients with early-onset myasthenia gravis.41 Cancer was not overrepre- sented as a cause
gravis and thymic hyper- plasia. Thyroiditis is the most of death in a Norwegian popu- lation-based study.36
common coexist- ing condition, followed by systemic Lymphomas have consistently been seen with a slightly
lupus erythe- matosus and rheumatoid arthritis. In increased frequency in patients with myasthenia
patients with ocular myasthenia, thyroid disease is gravis.42 Azathioprine used as immunosuppressive
espe- cially common. treatment for myas- thenia did not influence the
Myasthenia gravis occurs in one third of all general cancer risk in a Danish population study,43
patients with a thymoma. Although the strong whereas this treatment used for inflammatory
association between thymoma and myasthenia gravis bowel disease slightly increased the cancer risk in a
is unique, thymoma is also associated with an similar Dutch study,44 and the risk of lip cancer also
increased risk of certain other autoim- mune increased with high-dose azathioprine.45
disorders. Blood cytopenias, hypogamma- globulinemia,
Treatment for myasthenia gravis can increase
polymyositis, the POEMS syndrome (polyneuropathy,
the risk of coexisting disorders. Prednisolone
organomegaly, endocrinopathy, M component, and skin
necessitates prophylaxis against osteoporosis, and
changes), neuromyotonia, and autoimmune
patients should be monitored for weight gain,
encephalitis occur with an in- creased frequency among
elevations in blood glucose levels, and hy-
patients with a thymoma but are rare in patients with
pertension. Anticholinergic drugs for symptom- atic
myasthenia gravis. Neuromyelitis optica with
treatment have transient and dose-limiting effects on
aquaporin-4 anti- bodies has a prevalence of 40 cases
the autonomic nervous system.
per 1 million population,31 has a specific association Concomitant disease represents a major chal-
with myasthenia gravis, and can occur either before lenge in treating patients with myasthenia gra- vis. An
or after the onset of myasthenia gravis.32 Amyo- increasing number of patients are elderly, with
trophic lateral sclerosis (ALS) occurs in patients with reduced mobility, respiratory function, and quality of
myasthenia gravis more often than would be life due to the combined effects of several health
expected on the basis of the risk in the gen- eral issues.
population. Autoimmune disease in general

T HE R A p Y
represents a risk factor for ALS, but the association
with myasthenia gravis is especially strong.33,34 DRUGS FOR S YMPTOMATic THERAPY
Myocarditis is rare but occurs with an in-
creased frequency in patients with myasthenia gravis, All subgroups of myasthenia gravis respond to
as indicated by numerous single case series and acetylcholinesterase inhibition (Fig. 1). Pyridostig-
reports.35 However, myasthenia-related mine is the preferred drug for the treatment of
symptoms in all myasthenia gravis subgroups2,11,12

2574
N ENGL J MED 375;26 NEJM.ORG DECEMBER 29, 2016
M YASTHENIA G R AVIS

(Table 2). Neostigmine and ambenonium chloride are


ternate days. A major aim of treatment for ocu- lar
also inhibitors of acetylcholinesterase but are less
myasthenia gravis is to prevent generalization of the
effective than pyridostigmine in most pa- tients.
disease. Retrospective and observational studies
Increasing the release of acetylcholine
strongly indicate that prednisolone mono- therapy
presynaptically by administering 3,4-diamino-
reduces this risk. Low-dose glucocorti- coid treatment
pyridine or ephedrine usually has a mild benefi- cial
is therefore recommended by many experts for
effect, but it is rarely sufficient for practical use.
patients with ocular myasthenia gravis who have
Myasthenia gravis with muscle-specific ki- nase
persistent symptoms and risk factors such as
antibodies generally has a less favorable response
detectable acetylcholine receptor antibodies, an
to drugs administered for symptomatic therapy than
enlarged thymus,47 or results of neurophysiological
do the other disease subgroups.23 Juvenile
tests showing additional dis- ease involvement of
myasthenia gravis often has an excellent response to
nonocular muscles.13,48,49
pyridostigmine.15,16 The dose of pyri- dostigmine is
decided on the basis of the effect on muscle In most patients, azathioprine is added to
strength and dose-dependent side effects, most prednisolone because this combination provides a
frequently involving the gastroin- testinal tract. better functional result with fewer side effects than
Typical side effects are diarrhea, abdominal pain or prednisolone monotherapy.50 If glucocorti- coids are
cramps, increased f latus, nausea, and increased contraindicated or if the patient declines them,
salivation, as well as uri- nary urgency and increased azathioprine can be given alone. The rec- ommended
sweating. Most pa- tients are capable of adjusting dose is 2 to 3 mg per kilogram of body weight.
their own dose, with possible variation from day to Azathioprine inhibits purine syn- thesis and thus cell
day. The ef- fect of pyridostigmine remains proliferation, with a particu- larly strong effect on B
unchanged over a period of years. For patients who and T cells. Thiopurine methyltransferase activity
have mild disease and nearly full remission with should be tested before treatment, if the test is
symptom- atic therapy with drugs, no other drug available, because low activity increases the risk that
therapy is recommended (Fig. 3A). azathioprine will have toxic side effects.51,52 Enzyme
activity is ab- sent in only 0.3% of the general
IMMUNOSUPPRESSivE D RUG THERAPY population, whereas low enzyme activity is found
in up to 10% of the population, with some variation
Most patients with myasthenia gravis need immu- re- flecting genetic variants. Azathioprine is not rec-
nosuppressive medication to meet the treatment goals ommended in patients with no thiopurine methyl-
of full or nearly full physical function and high quality transferase activity and should be used with
of life. Immunosuppressive medica- tion is given to all caution and only at a low dose in patients with low
patients who do not have a fully satisfactory activity. The effect of azathioprine on myas- thenic
functional result with symptom- atic and supportive weakness often takes months to appear, and patients
therapy alone. Expert con- sensus and data from need to receive other immunosup- pressive
limited controlled trials support the use of medication during this period. Long- term treatment
prednisone or prednisolone in combination with is safe in all patients, including those who are
azathioprine as first-line treat- ment.11,12,46 Prednisone young.43
and prednisolone are re- garded as equally effective.
Most guidelines recommend mycophenolate
Alternate-day dosing, which is often used to reduce
mofetil for mild or moderate myasthenia gravis, even
the side effects of glucocorticoids, does not usually
though an additional benefit with this medi- cation
lead to unwant- ed disease fluctuations, but the
was not proven in two short-term pro- spective
evidence for re- duced side effects is weak.2 The
studies, which had methodologic limi- tations.53-55 The
dose is usually increased gradually (up to 60 to 80
drug blocks purine synthesis and interferes with B-
mg on alter- nate days) to avoid an initial
cell and T-cell proliferation. Methotrexate,
deterioration. After stable control of symptoms has
cyclosporine, and tacrolimus are al- ternative secondary
been achieved and the addition of other treatments
immunosuppressive drugs.56-58 The effect of these drugs
has further improved symptom control, the
is probably similar to that of azathioprine.
glucocorticoid dose should be slowly reduced to the
Rituximab represents a potentially potent treat-
lowest ef- fective level, which is often 10 to 40 mg
on al- ment for myasthenia gravis.59 This monoclonal

N ENGL J MED 375;26 NEJM.ORG DECEMBER 29, 2016 2575


25
76

N
NEJM.OR
2016

The New
England
Journal of
Medicine
Downloaded Table 2. Drugs Used Most Frequently for the Treatment of Myasthenia Gravis.
from Drug Mode of Action Dose Side Effects Risks and Contraindications
nejm.org at
TULANE Pyridostigmine Symptomatic; acetylcholinesterase Single dose: 10–120 mg; daily Cholinergic autonomic effects Cholinergic crisis
UNIV on inhibition dose: 40–600 mg
January 16, Prednisone or Immunomodulation Induction dose: 40–80 mg daily; Widespread dose-dependent glucocorticoid Gastrointestinal bleeding, cushingoid T
2019. For prednisolone stable dose: 5–20 mg daily; effects appearance he
personal use alternate-day treatment is an N
only. No alternative E
other uses Azathioprine Suppression of B and T cells 50–250 mg daily Nausea, vomiting, tiredness, infections, Leukopenia, liver toxicity W
without night sweats E
permission.
Mycophenolate mofetil Suppression of B and T cells 1.5–2 g daily Nausea, vomiting, diarrhea, joint pain, Leukopenia, progressive multifocal N
Copyright © infections, tiredness leukoencephalopathy; contraindi- cated GL
2016
during pregnancy
Massachusett A
s Medical Rituximab Suppression of B cells 0.5–1 g, repeated after 2 wk; can Nausea, infections, infusion-related Progressive multifocal leukoencepha- N
Society. All be repeated at 6-mo intervals problems lopathy
D
rights Methotrexate Inhibition of folate metabolism Gradual increase to 20 mg/wk Nausea, infections, lung disease Leukopenia, liver toxicity; contraindicat- JO
reserved. ed during pregnancy
U
Cyclosporine Suppression of T cells and natural 2.5–5 mg/kg of body weight daily Nausea, hypertension, infections, Kidney toxicity R
killer cells hypertrichosis
N
Tacrolimus Suppression of T cells and natural 3 mg daily Nausea, infections, lung disease, hyperten- Liver and kidney toxicity A
killer cells sion, neuropsychiatric problems
L
Cyclophosphamide Suppression of B and T cells 1–5 mg per kg administered by Nausea, vomiting, alopecia, discoloration Leukopenia of
intravenous infusion every 4 of nails and skin, infections M
wk for a limited period
Intravenous immune Suppression of B and T cells, 2 g per kg administered over a Nausea, headache, fever, hypotension or IgA deficiency, allergic reactions
globulin neutralization of autoantibodies period of 2 to 5 days hypertension, local skin reactions
M YASTHENIA G R AVIS

A Proposed Treatment for Generalized Myasthenia Gravis B Proposed Treatment for Severe Exacerbations of Generalized
Myasthenia Gravis

Intensive care
Diagnosis confirmed
IV immune globulin or plasma exchange
Treatment of infection and other
precipitating events

Acetylcholinesterase inhibitor and


thymectomy (if early onset or thymoma)
Intensify long-
Improvement? Yes term
immuno-
Continue with suppression
Clinical remission? Yes acetylcholin-
esterase inhibitor No

No
Plasma exchange or IV immune globulin
Glucocorticoids in megadose
Intensive care
Prednisolone and azathioprine

Intensify long-
Continue with Improvement? Yes term
Adequate effect? Yes lowest immuno-
possible dose suppression

No No

Mycophenolate mofetil for mild Rituximab


or moderate symptoms Intensive care
Rituximab for severe symptoms Treatment of complications
Other immunosuppressive drugs Other immunosuppressive drugs
Never give up

Continue
Sufficient effect? Yes
treatment

No

Other immunosuppressive drugs


(methotrexate, cyclosporine, tacrolimus)
Reevaluation of diagnosis
Off-label drugs

Figure 3. Proposed Treatment Algorithms for Generalized Myasthenia Gravis and for Severe Exacerbations of Generalized Disease. Panel
A shows treatments for generalized myasthenia gravis, and Panel B shows treatments for severe exacerbations. Both algorithms are from
Gilhus and Verschuuren.2 IV denotes intravenous.

antibody binds specifically to the CD20 surface


have a substantial improvement with rituximab.60 A
antigen on B lymphocytes and should therefore be
recommended induction dose has not been
effective in antibody-mediated diseases such as
established. The treatment should be repeated if the
myasthenia gravis. T-cell responses are also
symptoms recur after several months. Con- cerns
influenced by rituximab. A group of experts who
regarding rituximab are the risk of pre- cipitating
recently issued guidelines for the management of
additional autoimmune disorders and JC virus–related
myasthenia gravis could not reach a consensus on the
progressive multifocal leukoen- cephalopathy. The
role of rituximab.11 Evidence from small case series
treatment scheme for general- ized myasthenia
indicates that two thirds of patients with severe
gravis is summarized in Fig- ure 3A.
myasthenia gravis and an insuffi- cient response to
prednisolone and azathioprine A number of monoclonal antibody drugs have

N ENGL J MED 375;26 NEJM.ORG DECEMBER 29, 2016 2577

The New England Journal of Medicine


Downloaded from nejm.org at TULANE UNIV on January 16, 2019. For personal use only. No other uses without permission.
Copyright © 2016 Massachusetts Medical Society. All rights reserved.
T he NE W E NGL A ND JOU R NA L o f M E DICI NE

a proven effect in the treatment of other autoim- mune ing 3 years of observation. The differences were
disorders. They interfere with B cells, T cells, regarded as clinically meaningful. All thymic tissue
complement, or other immunoactive elements.61,62 needs to be removed, including the tissue embedded
Formal evidence and cost–benefit information are in mediastinal fat. Video- and robot- assisted methods
lacking for the use of these drugs in patients with minimize the surgical proce- dure, are preferred by
myasthenia gravis, although preliminary ob- servations most patients, and pro- vide the same benefit as
and mechanisms of drug action make several of them traditional open, transsternal thymectomy as long
promising alternatives. Autolo- gous hematopoietic as all tissue is removed.
stem-cell transplantation was recently reported to Guidelines and consensus statements recom- mend
provide stable and treatment- free remission in seven early thymectomy for patients with early- onset
patients.63 myasthenia gravis.2,11,12 These patients most often have
Patients with myasthenia gravis that develops late or thymic hyperplasia. Thymectomy should also be
is associated with thymoma or muscle- specific considered in children.68 Most patients with late-
kinase antibodies tend to have the most severe onset disease have an atrophic thymus.64 However,
disease and usually need long-term immu- thymic hyperplasia can occur in younger patients in
nosuppressive drug treatment, although some the late-onset subgroup. Thymectomy should also be
patients with late-onset myasthenia gravis have considered in patients with generalized myasthenia
disease that is milder and more similar to early- onset gravis who have acetylcholine receptor antibodies
disease. The presence of antibodies against muscle- and whose symptoms developed at the age of 50 to 65
specific kinase, titin, ryanodine receptor, or Kv1.4 is years,67 especially when the biomarkers show similari-
an indication for immunosuppres- sion. Myasthenia ties with early-onset disease. Current evidence does
gravis associated with muscle- specific kinase not support thymectomy in patients with
antibodies has a particularly fa- vorable response to myasthenia gravis and muscle-specific kinase or LRP4
rituximab. antibodies.11 Thymectomy is also not rec- ommended
for patients with ocular myasthenia, since there is
THYMEc TOMY insufficient evidence that surgery prevents
In patients with a thymoma and myasthenia gravis, generalization or results in remission. However, it has
thymectomy should be performed to re- move the been argued that thymectomy should be considered
tumor. A benefit after total thymec- tomy has been for the treatment of ocular myasthenia gravis when
reported for this subgroup; an even greater benefit drug treatment has failed, the patient has acetylcholine
of total thymectomy has been reported for patients receptor antibodies, and neurophysiological tests
with early-onset my- asthenia gravis without a indicate a risk of generalized disease.13
thymoma. The thymus has a key role in inducing Thymectomy is usually not recommended for
acetylcholine receptor antibody production in patients in whom all muscle antibody tests are
patients with myasthe- nia gravis.64 Many studies negative. However, some of these patients have
have compared the outcomes for patients who acetylcholine receptor antibodies that are not
undergo thymecto- my with the outcomes for those detected by routine assays. Therefore, in patients with
who do not, and nearly all the studies have shown negative muscle antibody tests who have
a better outcome in the thymectomy group. 65,66 A generalized disease with biomarkers similar to those
recent international, randomized, controlled trial involv- in patients with early-onset disease, thymec- tomy may
ing 126 patients with early-onset or late-onset be considered if the disease fails to respond to
myasthenia gravis confirmed a distinct benefit from immunosuppressive drugs.11
early thymectomy, supporting thymectomy in patients
with generalized disease, a disease duration of less MYASTHENiA GRAVIS CRisis
than 3 to 5 years, an age of less than 60 to 65 years,
and symptoms not fully relieved by Patients with worsening weakness who require
anticholinesterase drugs.67 Patients who underwent intubation or noninvasive ventilation should re- ceive
thymectomy, as compared with those who did not fast-acting immunosuppressive agents and intensive
receive surgical treatment, had significant reductions care. An impending myasthenic crisis with rapid
in symptoms, immunosup- pressive drug treatment, and worsening and severe weakness war-
exacerbations dur-

2578
N ENGL J MED 375;26 NEJM.ORG DECEMBER 29, 2016
M YASTHENIA G R AVIS

rants a similar intervention.69 The threshold for and this approach is preferable to withholding the
deciding to admit a patient to an intensive care unit drug altogether.
should be low. Increasing generalized weak- ness, Respiratory insufficiency due to diaphragmatic and
respiratory dysfunction, cardiac dysfunc- tion, severe intercostal muscle weakness is a major threat. Special
infection, and coexisting conditions are all relevant attention should be paid to respiratory function
factors to consider in making this decision. during any surgical procedure, including thymectomy, in a
Measures such as vital capacity and blood gas patient with myasthenia gravis. Optimal treatment of
levels have limited value, since deterioration can be all coexisting conditions is an important component of
rapid and unexpected as a result of the characteristic the management of myasthenia gravis. This can be a
myasthenic fatigability. Intravenous immune globulin particular chal- lenge in elderly patients with multiple
and plasma ex- change are regarded as equally coexisting conditions.
effective in treat- ing severe myasthenia gravis.70-72 Oral administration of pyridostigmine and
The choice be- tween them depends on individual prednisone or prednisolone is safe during preg-
patient factors and institutional experience, nancy.75,76 Current information indicates that
availability, and tradition. Intravenous immune treatment with azathioprine and cyclosporine is safe
globulin is often regarded as more convenient with as well. Mycophenolate mofetil and metho- trexate are
less severe side effects. A patient may have a contraindicated during pregnancy be- cause of
response to one treatment approach but not the teratogenic risks. Women are advised to avoid
other. The treatment effect is restricted to a period pregnancy for up to 1 year after finishing rituximab
of a few months and should therefore be combined treatment. Intravenous immune glob- ulin and plasma
with long-term immunosuppressive treatment. In some exchange are useful for worsen- ing weakness during
patients, the treatment response is delayed. Vig- orous pregnancy. Lactation should be encouraged. Transient
immunosuppressive treatment combined with intensive neonatal myasthenia occurs in 15% of children as a
care should be maintained as long as necessary to result of trans- placental IgG transfer of antibodies
induce remission. Myasthenic crisis with a need for against acetylcholine receptor, muscle-specific
respiratory support is now rare in patients with
kinase, or LRP4.76,77
myasthenia gravis, and mortality during myasthenic
crisis is also low.69 The treatment scheme for severe
exacerbations F U T U R E D I R E C T IONS
of myasthenia gravis is shown in Figure 3B.
With specialized treatment, the great majority
SuPPORTivE THERAPY AND MANAGEMENT of patients with myasthenia gravis do well. They are
able to perform daily tasks and maintain a near-
Physical activity and systematic training pro- grams normal quality of life. However, only a few patients
at a low or medium level of intensity should be have a full remission, and most do not even have a
recommended for patients with myas- thenia gravis full pharmacologic remission. Al- though the
and tailored to the individual pa- tient.2,11,12 Overweight disease-inducing antibodies have been characterized in
should be avoided. Assistive devices can be helpful with detail, the treatment is far from immunospecific. Data
ocular symptoms.13 Muscle relaxants, penicillamine, from prospective, blinded, controlled studies comparing
and some antibiotics (f luoroquinolones, macrolides, treatments are lack- ing, and there have been few
and aminoglycosides) should be avoided, if possible, in well-controlled studies of individual drugs and
patients with myasthenia gravis. Statins can aggravate nondrug inter- ventions. Apart from paraneoplasia
and unmask myasthenia gravis, but the presence of associated with thymoma, the causes of myasthenia
myasthenia gravis is not regarded as a gravis are unknown.
contraindication if statins are needed, and the Monoclonal antibodies have selective binding
indications for statin treatment in patients with and a high specificity regarding immunologic actions
myasthenia gravis are the same as the indications for but do not necessarily have any specific- ity for the
such treatment in patients without myasthe- nia treatment of myasthenia gravis. Ongo- ing trials are
gravis.73,74 If a drug appears to be indicated, vigilance evaluating more targeted immu- noactive therapy.
in looking for worsening of weakness is important Antigen-specific treatment is
when the new drug is introduced,

N ENGL J MED 375;26 NEJM.ORG DECEMBER 29, 2016 2579


T he NE W E NGL A ND JOU R NA L o f M E DICI NE

being developed for myasthenia gravis associat- ed


with acetylcholine receptor, muscle-specific kinase,
and LRP4 antibodies, through interac- tion with myasthenia gravis show great variation within and
regulatory B or T cells.7,78,79 between countries. Implementing best-prac- tice
Even with today’s knowledge and available standards universally represents a major chal- lenge.
treatments, it is a challenge to find the optimal This is especially important because my- asthenia
treatment for the individual patient. Specialized gravis is a potentially reversible disorder with
diagnostic procedures and expert follow-up over time treatment options that can make a huge difference
improve treatment results. Standards and for the patient.
No potential conflict of interest relevant to this article was
possibilities for the diagnosis and treatment of
reported.

Disclosure forms provided by the author are available with the


full text of this article at NEJM.org.

R EFEREN c ES
1. Gilhus NE, Skeie GO, Romi F, Lazari-
dis K, Zisimopoulou P, Tzartos S. Myas- thenia gravis — 12. Skeie GO, Apostolski S, Evoli A, et al. 24. Hong Y, Li HF, Skeie GO, et al. Auto-
autoantibody character- istics and their implications for Guidelines for treatment of autoimmune antibody profile and clinical characteris- tics
therapy. Nat Rev Neurol 2016;12:259-68. neuromuscular transmission disorders. Eur in a cohort of Chinese adult myasthe- nia
2. Gilhus NE, Verschuuren JJ. Myasthe- nia J Neurol 2010;17:893-902. gravis patients. J Neuroimmunol 2016;
gravis: subgroup classification and therapeutic 13. Kerty E, Elsais A, Argov Z, Evoli A, 298:51-7.
strategies. Lancet Neurol 2015; 14:1023-36. Gilhus NE. EFNS/ENS guidelines for the 25. Higuchi O, Hamuro J, Motomura M,
3. Querol L, Illa I. Myasthenia gravis and treatment of ocular myasthenia. Eur J Yamanashi Y. Autoantibodies to low-den-
the neuromuscular junction. Curr Opin Neurol 2014;21:687-93. sity lipoprotein receptor-related protein 4 in
Neurol 2013;26:459-65. 14. Heldal AT, Owe JF, Gilhus NE, Romi F. myasthenia gravis. Ann Neurol 2011;
4. Carr AS, Cardwell CR, McCarron PO, Seropositive myasthenia gravis: a nation- 69:418-22.
McConville J. A systematic review of pop- wide epidemiologic study. Neurology 2009; 26. Zhang B, Tzartos JS, Belimezi M, et al.
ulation based epidemiological studies in 73:150-1. Autoantibodies to lipoprotein-related pro-
myasthenia gravis. BMC Neurol 2010;10: 46. 15. VanderPluym J, Vajsar J, Jacob FD, tein 4 in patients with double-seronega-
5. Verschuuren J, Strijbos E, Vincent A. Mah JK, Grenier D, Kolski H. Clinical tive myasthenia gravis. Arch Neurol 2012;
Neuromuscular junction disorders. Handb Clin characteristics of pediatric myasthenia: a 69:445-51.
Neurol 2016;133:447-66. surveillance study. Pediatrics 2013;132(4): 27. Leite MI, Jacob S, Viegas S, et al. IgG1
6. Zisimopoulou P, Brenner T, Trakas N, e939-e944. antibodies to acetylcholine receptors in
Tzartos SJ. Serological diagnostics in my- asthenia gravis 16. Liew WKM, Kang PB. Update on juve- ‘seronegative’ myasthenia gravis. Brain
based on novel assays and recently identified antigens. nile myasthenia gravis. Curr Opin Pediatr 2008;131:1940-52.
Autoimmun Rev 2013;12:924-30. 2013;25:694-700. 28. Gasperi C, Melms A, Schoser B, et al.
7. Huijbers MG, Lipka AF, Plomp JJ, Niks 17. Renton AE, Pliner HA, Provenzano C, et Anti-agrin autoantibodies in myasthenia
EH, van der Maarel SM, Verschuuren JJ. Pathogenic al. A genome-wide association study of gravis. Neurology 2014;82:1976-83.
immune mechanisms at the neuromuscular synapse: myasthenia gravis. JAMA Neurol 2015;72: 29. Gallardo E, Martínez-Hernández E,
the role of specific antibody-binding epitopes in my- 396-404. Titulaer MJ, et al. Cortactin autoantibod- ies
asthenia gravis. J Intern Med 2014;275: 12-26. 18. Saruhan-Direskeneli G, Hughes T, in myasthenia gravis. Autoimmun Rev
8. Chiou-Tan FY, Gilchrist JM. Repetitive Yilmaz V, et al. Genetic heterogeneity 2014;13:1003-7.
nerve stimulation and single-fiber electro- within the HLA region in three distinct 30. Gilhus NE, Nacu A, Andersen JB, Owe JF.
myography in the evaluation of patients clinical subgroups of myasthenia gravis. Clin Myasthenia gravis and risks for comor-
with suspected myasthenia gravis or Lam- Immunol 2016;166-167:81-8. bidity. Eur J Neurol 2015;22:17-23.
bert-Eaton myasthenic syndrome: review of 19. Nacu A, Andersen JB, Lisnic V, Owe JF, 31. Flanagan EP, Cabre P, Weinshenker
recent literature. Muscle Nerve 2015; Gilhus NE. Complicating autoimmune BG, et al. Epidemiology of aquaporin-4
52:455-62. diseases in myasthenia gravis: a review. autoimmunity and neuromyelitis optica
9. Jordan B, Kellner J, Jordan K, Bähre M, Autoimmunity 2015;48:362-8. spectrum. Ann Neurol 2016;79:775-83.
Behrmann C, Zierz S. Thymic pathologies in 20. Stergiou C, Lazaridis K, Zouvelou V, et 32. Freitas E, Guimarães J. Neuromyelitis
myasthenia gravis: a preoperative assess- al. Titin antibodies in “seronegative” optica spectrum disorders associated with
ment of CAT scan and nuclear based im- myasthenia gravis — a new role for an old other autoimmune diseases. Rheumatol Int
aging. J Neurol 2016;263:641-8. antigen. J Neuroimmunol 2016;292:108- 2015;35:243-53.
10. Romi F, Skeie GO, Gilhus NE, Aarli JA. 15. 33. Gotaas HT, Skeie GO, Gilhus NE. My-
Striational antibodies in myasthenia gravis: reactivity 21. Romi F, Suzuki S, Suzuki N, Petzold A, asthenia gravis and amyotrophic lateral
and possible clinical sig- nificance. Arch Neurol Plant GT, Gilhus NE. Anti-voltage-gated sclerosis: a pathogenic overlap. Neuro-
2005;62:442-6. potassium channel Kv1.4 antibodies in muscul Disord 2016;26:337-41.
11. Sanders DB, Wolfe GI, Benatar M, et al. myasthenia gravis. J Neurol 2012;259: 34. Staff NP, Appel SH. The immune sys-
International consensus guidance for man- agement of 1312-6. tem continues to knock at the ALS door.
myasthenia gravis: executive summary. Neurology 22. Suzuki S, Baba A, Kaida K, et al. Car- Neuromuscul Disord 2016;26:335-6.
2016;87:419-25. diac involvements in myasthenia gravis 35. Mavrogeni S, Ntoskas T, Gialafos E, et
associated with anti-Kv1.4 antibodies. Eur J al. Silent myocarditis in myasthenia
Neurol 2014;21:223-30. gravis: role of cardiovascular magnetic
23. Guptill JT, Sanders DB, Evoli A. Anti- resonance imaging. Int J Cardiol 2016;
MuSK antibody myasthenia gravis: clini- cal 202:629-30.
findings and response to treatment in two 36. Owe JF, Daltveit AK, Gilhus NE. Causes
large cohorts. Muscle Nerve 2011;44: 36-40. of death among patients with my- asthenia
gravis in Norway between 1951

2580
N
ENGL J
MED
375;26
NEJM.
ORG
DECEM
BER
29,
2016
M YASTHENIA G R AVIS

and 2001. J Neurol Neurosurg Psychiatry


2006;77:203-7. myasthenia gravis. Neurology 1998;50: myasthenia gravis (an evidence-based re-
1778-83. view): report of the Quality Standards
37. Furlund Owe J, Skulstad Davidsen E,
Subcommittee of the American Academy of
Eide GE, Gerdts E, Gilhus NE. Left ven-
51. Rae W, Burke G, Pinto A. A study of Neurology. Neurology 2000;55:7-15.
tricular long-axis function in myasthenia
the utility of azathioprine metabolite test-
gravis. J Neurol 2008;255:1777-84.
ing in myasthenia gravis. J Neuroimmu- nol 66. Cea G, Benatar M, Verdugo RJ, Salinas
38. Suzuki S, Utsugisawa K, Yoshikawa H, et
2016;293:82-5. RA. Thymectomy for non-thymomatous
al. Autoimmune targets of heart and
52. Sanderson JD. TPMT testing before myasthenia gravis. Cochrane Database Syst
skeletal muscles in myasthenia gravis.
starting azathioprine or mercaptopurine: Rev 2013;10:CD008111.
Arch Neurol 2009;66:1334-8.
surely just do it? Gastroenterology 2015; 67. Wolfe GI, Kaminski HJ, Aban IB, et al.
39. Pedersen EG, Pottegård A, Hallas J, et
149:850-3. Randomized trial of thymectomy in myas-
al. Myasthenia and risk of cancer: a
53. Burns TM, Sanders DB, Kaminski HJ, thenia gravis. N Engl J Med 2016;375:511-
population-based case-control study. Eur J
Wolfe GI, Narayanaswami P, Venitz J. Two 22.
Neurol 2014;21:773-8.
steps forward, one step back: mycopheno- 68. Ashfaq A, Bernes SM, Weidler EM,
40. Liu CJ, Chang YS, Teng CJ, et al. Risk of
late mofetil treatment for myasthenia gra- Notrica DM. Outcomes of thoracoscopic
extrathymic cancer in patients with
vis in the United States. Muscle Nerve thymectomy in patients with juvenile my-
myasthenia gravis in Taiwan: a nation-
2015;51:635-7. asthenia gravis. J Pediatr Surg 2016;51:
wide population-based study. Eur J Neurol
54. Sanders DB, Hart IK, Mantegazza R, et 1078-83.
2012;19:746-51.
al. An international, phase III, random- ized 69. Alshekhlee A, Miles JD, Katirji B,
41. Owe JF, Cvancarova M, Romi F, Gil-
trial of mycophenolate mofetil in my- Preston DC, Kaminski HJ. Incidence and
hus NE. Extrathymic malignancies in thy-
asthenia gravis. Neurology 2008;71:400-6. mortality rates of myasthenia gravis and
moma patients with and without myas-
55. Muscle Study Group. A trial of myco- myasthenic crisis in US hospitals. Neurol-
thenia gravis. J Neurol Sci 2010;290:66-9.
phenolate mofetil with prednisone as ini- ogy 2009;72:1548-54.
42. Yeh J-H, Lin C-C, Chen Y-K, Sung F-C,
tial immunotherapy in myasthenia gravis. 70. Gajdos P, Chevret S, Toyka KV. Intra-
Chiu H-C, Kao C-H. Excessive risk of can- cer
Neurology 2008;71:394-9. venous immunoglobulin for myasthenia
and in particular lymphoid malignancy in
56. Pasnoor M, He J, Herbelin L, et al. A gravis. Cochrane Database Syst Rev 2012;
myasthenia gravis patients: a population- based
randomized controlled trial of metho- 12:CD002277.
cohort study. Neuromuscul Disord
trexate for patients with generalized my- 71. Gajdos P, Chevret S, Clair B, Tranchant
2014;24:245-9.
asthenia gravis. Neurology 2016;87:57-64. C, Chastang C. Clinical trial of plasma ex-
43. Pedersen EG, Pottegård A, Hallas J, et
57. Yoshikawa H, Kiuchi T, Saida T, Taka- change and high-dose intravenous immu-
al. Risk of non-melanoma skin cancer in
mori M. Randomised, double-blind, pla- noglobulin in myasthenia gravis. Ann
myasthenia patients treated with aza-
cebo-controlled study of tacrolimus in Neurol 1997;41:789-96.
thioprine. Eur J Neurol 2014;21:454-8.
myasthenia gravis. J Neurol Neurosurg 72. Barth D, Nabavi Nouri M, Ng E, Nwe P,
44. van den Heuvel TRA, Wintjens DSJ,
Psychiatry 2011;82:970-7. Bril V. Comparison of IVIg and PLEX in
Jeuring SFG, et al. Inflammatory bowel
58. Tindall RSA, Phillips JT, Rollins JA, patients with myasthenia gravis. Neurol-
disease, cancer and medication: cancer
Wells L, Hall K. A clinical therapeutic trial ogy 2011;76:2017-23.
risk in the Dutch population-based IBDSL
of cyclosporine in myasthenia gravis. Ann N 73. Argov Z. Statins and the neuromus-
cohort. Int J Cancer 2016;139:1270-80.
Y Acad Sci 1993;681:539-51. cular system: a neurologist’s perspective.
45. Na R, Laaksonen MA, Grulich AE, et al.
59. Keung B, Robeson KR, DiCapua DB, et Eur J Neurol 2015;22:31-6.
High azathioprine dose and lip cancer risk in
al. Long-term benefit of rituximab in 74. Gilhus NE. Is it safe to use statins in
liver, heart, and lung transplant recipi- ents:
MuSK autoantibody myasthenia gravis patients with myasthenia gravis? Nat Clin
a population-based cohort study. J Am Acad
patients. J Neurol Neurosurg Psychiatry Pract Neurol 2009;5:8-9.
Dermatol 2016;74(6):1144-1152.e6.
2013;84:1407-9. 75. Norwood F, Dhanjal M, Hill M, et al.
46. Hart IK, Sathasivam S, Sharshar T.
60. Iorio R, Damato V, Alboini PE, Evoli A. Myasthenia in pregnancy: best practice
Immunosuppressive agents for myasthe- nia
Efficacy and safety of rituximab for myas- guidelines from a U.K. multispecialty
gravis. Cochrane Database Syst Rev
thenia gravis: a systematic review and working group. J Neurol Neurosurg Psy-
2007;4:CD005224.
meta-analysis. J Neurol 2015;262:1115-9. chiatry 2014;85:538-43.
47. Binks S, Vincent A, Palace J. Myasthe-
61. Reichert JM. Antibodies to watch in 76. Ferrero S, Pretta S, Nicoletti A, Petrera
nia gravis: a clinical-immunological up-
2016. MAbs 2016;8:197-204. P, Ragni N. Myasthenia gravis: manage-
date. J Neurol 2016;263:826-34.
62. Milo R. Therapeutic strategies target- ment issues during pregnancy. Eur J Ob- stet
48. Benatar M, Kaminski H. Medical and
ing B-cells in multiple sclerosis. Autoim- Gynecol Reprod Biol 2005;121:129-38.
surgical treatment for ocular myasthenia.
mun Rev 2016;15:714-8. 77. Hoff JM, Daltveit AK, Gilhus NE. My-
Cochrane Database Syst Rev 2012;12:
63. Bryant A, Atkins H, Pringle CE, et al. asthenia gravis in pregnancy and birth:
CD005081.
Myasthenia gravis treated with autologous identifying risk factors, optimising care.
49. Benatar M, Mcdermott MP, Sanders DB,
hematopoietic stem cell transplantation. Eur J Neurol 2007;14:38-43.
et al. Efficacy of Prednisone for the Treat-
JAMA Neurol 2016;73:652-8. 78. Steinman L. The road not taken: anti-
ment of Ocular Myasthenia (EPITOME): a
64. Marx A, Pfister F, Schalke B, Saruhan- gen-specific therapy and neuroinflamma-
randomized, controlled trial. Muscle
Direskeneli G, Melms A, Ströbel P. The tory disease. JAMA Neurol 2013;70:1100-1.
Nerve 2016;53:363-9.
different roles of the thymus in the patho- 79. Dalakas MC. Future perspectives in
50. Palace J, Newsom-Davis J, Lecky B. A
genesis of the various myasthenia gravis target-specific immunotherapies of myas-
randomized double-blind trial of pred-
subtypes. Autoimmun Rev 2013;12:875-84. thenia gravis. Ther Adv Neurol Disord
nisolone alone or with azathioprine in
65. Gronseth GS, Barohn RJ. Practice 2015;8:316-27.
parameter: thymectomy for autoimmune Copyright © 2016 Massachusetts Medical Society.

N ENGL J MED 375;26 NEJM.ORG DECEMBER 29, 2016 2581

Anda mungkin juga menyukai